Structure-Based Discovery of A2A Adenosine Receptor Ligands
Abstract

The recent determination of X-ray structures of pharmacologically relevant GPCRs has made these targets accessible to structure-based ligand discovery. Here we explore whether novel chemotypes may be discovered for the A2A adenosine receptor, based on complementarity to its recently determined structure. The A2A adenosine receptor signals in the periphery and the CNS, with agonists explored as anti-inflammatory drugs and antagonists explored for neurodegenerative diseases. We used molecular docking to screen a 1.4 million compound database against the X-ray structure computationally and tested 20 high-ranking, previously unknown molecules experimentally. Of these 35% showed substantial activity with affinities between 200 nM and 9 μM. For the most potent of these new inhibitors, over 50-fold specificity was observed for the A2A versus the related A1 and A3 subtypes. These high hit rates and affinities at least partly reflect the bias of commercial libraries toward GPCR-like chemotypes, an issue that we attempt to investigate quantitatively. Despite this bias, many of the most potent new ligands were novel, dissimilar from known ligands, providing new lead structures for modulation of this medically important target.
Introduction
Abbreviations: GPCR, G-protein-coupled receptor; AR, adenosine receptor; CNS, central nervous system; SEA, similarity ensemble approach; PDB, Protein Data Bank; WOMBAT, World of Molecular Bioactivity; CHO, Chinese hamster ovary; DMEM, Dulbecco’s modified Eagle medium; PKA, protein kinase A; DLS, dynamic light scattering.
Chart 1

Figure 1

Figure 1. Binding mode of the cocrystallized ligand 6 (A) and the predicted binding modes of the seven ligands discovered in the docking screen (B−H). The A2A AR binding site is shown in white ribbons with the side chains of Glu169 and Asn253 in sticks. In (A) the cocrystallized ligand 6 is shown using orange carbon atoms. In (B−H), the crystallographic ligand is shown using blue lines and the docking poses for the ligands are depicted with orange carbon atoms. Black dotted lines indicate hydrogen bonds. The compounds are (B) 7, (C) 8, (D) 9, (E) 10, (F) 11, (G) 12, and (H) 13.
Methods
Preparation of the Molecular Docking Screen
Similarity and Library Bias Calculations
A2A AR Receptor Binding and Functional Assay
Cell Culture and Membrane Preparation
Binding Assays
Cyclic AMP Accumulation Assay
Counterscreen for Colloidal Inhibition
Results and Discussion
Molecular Docking Screen and Compound Selection
![]() |
Radioligand Displacement Assays and Docking Hit Rate
Figure 2

Figure 2. Representative dose−response curves for displacement of binding of the radiolabeled A2A AR agonist 3 by compounds 9, 10, and 11.
![]() |
Measured in three independent experiments.
representative DOCK screen | |||
---|---|---|---|
target | no. of ZINC molecules similar to known ligandsa | hit rateb (%) | best potencyc (nM) |
adenosine receptors | 4240 | 35 | 200 |
adrenergic receptors | 4146 | 24 | 9 (19) |
adenylyl cyclases | 565 | 4 | 50000 (55) |
AmpC β-lactamase | 545 | 2−5 | 26000 (29) |
ZINC leadlike molecules with at least 10−10P values to annotated target ligands in WOMBAT using the similarity ensemble approach (SEA).
(Number of true ligands)/(number of predictions tested experimentally).
The affinity of the ligand with the best potency from the docking screen.
Predicted Binding Modes, Novelty, and Efficacy of the Discovered A2A AR Ligands
Figure 3

Figure 3. Functional assay based on measuring the production of cAMP for 3 (control), a potent A2A AR agonist, with or without 10 μM 9 or 11. The dose−response curve is shifted for both compounds, as expected in the case of competitive antagonistic inhibition. The % activation refers to production of cAMP normalized to the effect of 3 at 100 μM.
Is There Library Bias toward GPCR Chemotypes in Chemical Libraries?
Note Added after Initial Review of This Paper
Supporting Information
Table S1 of structures of the 500 top-ranking molecules from the docking screen. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
This work is supported by NIH Grant GM59957 (to B.K.S.), NIDDK Intramural Research Program (to K.A.J.), and a fellowship from the Knut and Alice Wallenberg Foundation (to J.C.). We thank A. Doak for aggregation assays and members of the Shoichet lab for docking “hit-list” evaluation. We thank Tudor Oprea for access to the WOMBAT database and John Overington for a prerelease version of the ChEMBL database.
References
This article references 56 other publications.
- 1Overington, J. P.; Al-Lazikani, B.; Hopkins, A. L. How many drug targets are there? Nat. Rev. Drug Discovery 2006, 5, 993– 996Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1Kju7jM&md5=14b5716f4e10b57ae71bf67e52a21929How many drug targets are there?Overington, John P.; Al-Lazikani, Bissan; Hopkins, Andrew L.Nature Reviews Drug Discovery (2006), 5 (12), 993-996CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. For the past decade, the no. of mol. targets for approved drugs has been debated. Here, we reconcile apparently contradictory previous reports into a comprehensive survey, and propose a consensus no. of current drug targets for all classes of approved therapeutic drugs. One striking feature is the relatively const. historical rate of target innovation (the rate at which drugs against new targets are launched); however, the rate of developing drugs against new families is significantly lower. The recent approval of drugs that target protein kinases highlights two addnl. trends: an emerging realization of the importance of polypharmacol., and also the power of a gene-family-led approach in generating novel and important therapies.
- 2Cherezov, V.; Rosenbaum, D. M.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Kuhn, P.; Weis, W. I.; Kobilka, B. K.; Stevens, R. C. High-resolution crystal structure of an engineered human beta(2)-adrenergic G protein-coupled receptor Science 2007, 318, 1258– 1265Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlGmur7I&md5=12c5bacb8464a4b243fe9341192b5b3bHigh-Resolution Crystal Structure of an Engineered Human β2-Adrenergic G Protein-Coupled ReceptorCherezov, Vadim; Rosenbaum, Daniel M.; Hanson, Michael A.; Rasmussen, Soren G. F.; Thian, Foon Sun; Kobilka, Tong Sun; Choi, Hee-Jung; Kuhn, Peter; Weis, William I.; Kobilka, Brian K.; Stevens, Raymond C.; Takeda, S.; Kadowaki, S.; Haga, T.; Takaesu, H.; Mitaku, S.; Fredriksson, R.; Lagerstrom, M. C.; Lundin, L. G.; Schioth, H. B.; Pierce, K. L.; Premont, R. T.; Lefkowitz, R. J.; Lefkowitz, R. J.; Shenoy, S. K.; Rosenbaum, D. M.Science (Washington, DC, United States) (2007), 318 (5854), 1258-1265CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Second extracellular loop, which in the β2-adrenergic receptor contains an unusual pair of disulfide bonds and an extra helix. This loop and the absence Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors constitute the largest family of eukaryotic signal transduction proteins that communicate across the membrane. We report the crystal structure of a human β2-adrenergic receptor-T4 lysozyme fusion protein bound to the partial inverse agonist carazolol at 2.4 angstrom resoln. The structure provides a high-resoln. view of a human G protein-coupled receptor bound to a diffusible ligand. Ligand-binding site accessibility is enabled by the second extracellular loop, which is held out of the binding cavity by a pair of closely spaced disulfide bridges and a short helical segment within the loop. Cholesterol, a necessary component for crystn., mediates an intriguing parallel assocn. of receptor mols. in the crystal lattice. Although the location of carazolol in the β2-adrenergic receptor is very similar to that of retinal in rhodopsin, structural differences in the ligand-binding site and other regions highlight the challenges in using rhodopsin as a template model for this large receptor family.
- 3Rosenbaum, D. M.; Cherezov, V.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Yao, X. J.; Weis, W. I.; Stevens, R. C.; Kobilka, B. K. GPCR engineering yields high-resolution structural insights into beta(2)-adrenergic receptor function Science 2007, 318, 1266– 1273Google ScholarThere is no corresponding record for this reference.
- 4Warne, T.; Serrano-Vega, M. J.; Baker, J. G.; Moukhametzianov, R.; Edwards, P. C.; Henderson, R.; Leslie, A. G. W.; Tate, C. G.; Schertler, G. F. X. Structure of a beta(1)-adrenergic G-protein-coupled receptor Nature 2008, 454, 486– 491Google ScholarThere is no corresponding record for this reference.
- 5Jaakola, V. P.; Griffith, M. T.; Hanson, M. A.; Cherezov, V.; Chien, E. Y. T.; Lane, J. R.; IJzerman, A. P.; Stevens, R. C. The 2.6 angstrom crystal structure of a human A(2A) adenosine receptor bound to an antagonist Science 2008, 322, 1211– 1217Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlyqtbfN&md5=5bdb862b41f345c244f3c162e058206bThe 2.6 Angstrom Crystal Structure of a Human A2A Adenosine Receptor Bound to an AntagonistJaakola, Veli-Pekka; Griffith, Mark T.; Hanson, Michael A.; Cherezov, Vadim; Chien, Ellen Y. T.; Lane, J. Robert; IJzerman, Adriaan P.; Stevens, Raymond C.Science (Washington, DC, United States) (2008), 322 (5905), 1211-1217CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The adenosine class of heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) mediates the important role of extracellular adenosine in many physiol. processes and is antagonized by caffeine. The authors have detd. the crystal structure of the human A2A adenosine receptor, in complex with a high-affinity subtype-selective antagonist, ZM241385, to 2.6 angstrom resoln. Four disulfide bridges in the extracellular domain, combined with a subtle repacking of the transmembrane helixes relative to the adrenergic and rhodopsin receptor structures, define a pocket distinct from that of other structurally detd. GPCRs. The arrangement allows for the binding of the antagonist in an extended conformation, perpendicular to the membrane plane. The binding site highlights an integral role for the extracellular loops, together with the helical core, in ligand recognition by this class of GPCRs and suggests a role for ZM241385 in restricting the movement of a tryptophan residue important in the activation mechanism of the class A receptors.
- 6Congreve, M.; Marshall, F. The impact of GPCR structures on pharmacology and structure-based drug design Br. J. Pharmacol. 2010, 159, 986– 996Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1ektro%253D&md5=e9ab946b907eb44edf516eb3231e6931The impact of GPCR structures on pharmacology and structure-based drug designCongreve, Miles; Marshall, FionaBritish Journal of Pharmacology (2010), 159 (5), 986-996CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)A review. After many years of effort, recent tech. breakthroughs have enabled the X-ray crystal structures of three G-protein-coupled receptors (GPCRs) (β1 and β2 adrenergic and adenosine A2a) to be solved in addn. to rhodopsin. GPCRs, like other membrane proteins, have lagged behind sol. drug targets such as kinases and proteases in the no. of structures available and the level of understanding of these targets and their interaction with drugs. The availability of increasing nos. of structures of GPCRs is set to greatly increase our understanding of some of the key issues in GPCR biol. In particular, what constitutes the different receptor conformations that are involved in signaling and the mol. changes which occur upon receptor activation. How future GPCR structures might alter our views on areas such as agonist-directed signaling and allosteric regulation as well as dimerization is discussed. Knowledge of crystal structures in complex with small mols. will enable techniques in drug discovery and design, which have previously only been applied to sol. targets, to now be used for GPCR targets. These methods include structure-based drug design, virtual screening and fragment screening. This review considers how these methods have been used to address problems in drug discovery for kinase and protease targets and therefore how such methods are likely to impact GPCR drug discovery in the future.
- 7Moro, S.; Gao, Z. G.; Jacobson, K. A.; Spalluto, G. Progress in the pursuit of therapeutic adenosine receptor antagonists Med. Res. Rev. 2006, 26, 131– 159Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XislOksLw%253D&md5=48f7a603f3c4044b2c48aba13a031e07Progress in the pursuit of therapeutic Adenosine receptor antagonistsMoro, Stefano; Gao, Zhan-Guo; Jacobson, Kenneth A.; Spalluto, GiampieroMedicinal Research Reviews (2006), 26 (2), 131-159CODEN: MRREDD; ISSN:0198-6325. (John Wiley & Sons, Inc.)A review. Ever since the discovery of the hypotensive and bradycardiac effects of adenosine, adenosine receptors continue to represent promising drug targets. First, this is due to the fact that the receptors are expressed in a large variety of tissues. In particular, the actions of adenosine (or methylxanthine antagonists) in the central nervous system, in the circulation, on immune cells, and on other tissues can be beneficial in certain disorders. Second, there exists a large no. of ligands, which have been generated by introducing several modifications in the structure of the lead compds. (adenosine and methylxanthine), some of them highly specific. Four adenosine receptor subtypes (A1, A2A, A2B, and A3) have been cloned and pharmacol. characterized, all of which are G protein-coupled receptors. Adenosine receptors can be distinguished according to their preferred mechanism of signal transduction: A1 and A3 receptors interact with pertussis toxin-sensitive G proteins of the Gi and Go family; the canonical signaling mechanism of the A2A and of the A2B receptors is stimulation of adenylyl cyclase via Gs proteins. In addn. to the coupling to adenylyl cyclase, all four subtypes may pos. couple to phospholipase C via different G protein subunits. The development of new ligands, in particular, potent and selective antagonists, for all subtypes of adenosine receptors has so far been directed by traditional medicinal chem. The availability of genetic information promises to facilitate understanding of the drug-receptor interaction leading to the rational design of a potentially therapeutically important class of drugs. Moreover, mol. modeling may further rationalize obsd. interactions between the receptors and their ligands. In this review, we will summarize the most relevant progress in developing new therapeutic adenosine receptor antagonists.
- 8Jacobson, K. A.; Gao, Z. G. Adenosine receptors as therapeutic targets Nat. Rev. Drug Discovery 2006, 5, 247– 264Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhvFOlsr4%253D&md5=85ad553154bd61f24a6bb594f960f9bcAdenosine receptors as therapeutic targetsJacobson, Kenneth A.; Gao, Zhan-GuoNature Reviews Drug Discovery (2006), 5 (3), 247-264CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Adenosine receptors are major targets of caffeine, the most commonly consumed drug in the world. There is growing evidence that they could also be promising therapeutic targets in a wide range of conditions, including cerebral and cardiac ischemic diseases, sleep disorders, immune and inflammatory disorders and cancer. After more than three decades of medicinal chem. research, a considerable no. of selective agonists and antagonists of adenosine receptors have been discovered, and some have been clin. evaluated, although none has yet received regulatory approval. However, recent advances in the understanding of the roles of the various adenosine receptor subtypes, and in the development of selective and potent ligands, as discussed in this review, have brought the goal of therapeutic application of adenosine receptor modulators considerably closer.
- 9Sebastiao, A. M.; Ribeiro, J. A. Adenosine receptors and the central nervous system Handb. Exp. Pharmacol. 2009, 471– 534Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sktr3N&md5=f1c88cee280ebf7783da9efe18eb099eAdenosine receptors and the central nervous systemSebastiao, Ana M.; Ribeiro, Joaquim A.Handbook of Experimental Pharmacology (2009), 193 (Adenosine Receptors in Health and Disease), 471-534CODEN: HEPHD2; ISSN:0171-2004. (Springer GmbH)A review. The adenosine receptors (ARs) in the nervous system act as a kind of "go-between" to regulate the release of neurotransmitters (this includes all known neurotransmitters) and the action of neuromodulators (e.g., neuropeptides, neurotrophic factors). Receptor-receptor interactions and AR-transporter interplay occur as part of the adenosine's attempt to control synaptic transmission. A2AARs are more abundant in the striatum and A1ARs in the hippocampus, but both receptors interfere with the efficiency and plasticity-regulated synaptic transmission in most brain areas. The omnipresence of adenosine and A2A and A1 ARs in all nervous system cells (neurons and glia), together with the intensive release of adenosine following insults, makes adenosine a kind of "maestro" of the tripartite synapse in the homeostatic coordination of the brain function. Under physiol. conditions, both A2A and A1 ARs play an important role in sleep and arousal, cognition, memory and learning, whereas under pathol. conditions (e.g., Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, stroke, epilepsy, drug addiction, pain, schizophrenia, depression), ARs operate a time/circumstance window where in some circumstances A1AR agonists may predominate as early neuroprotectors, and in other circumstances A2AAR antagonists may alter the outcomes of some of the pathol. deficiencies. In some circumstances, and depending on the therapeutic window, the use of A2AAR agonists may be initially beneficial; however, at later time points, the use of A2AAR antagonists proved beneficial in several pathologies. Since selective ligands for A1 and A2A ARs are now entering clin. trials, the time has come to det. the role of these receptors in neurol. and psychiatric diseases and identify therapies that will alter the outcomes of these diseases, therefore providing a hopeful future for the patients who suffer from these diseases.
- 10Blackburn, M. R.; Vance, C. O.; Morschl, E.; Wilson, C. N. Adenosine receptors and inflammation Handb. Exp. Pharmacol. 2009, 215– 269Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sktr%252FK&md5=266c99602d3e0ee0dcfc81a4dfefef47Adenosine receptors and inflammationBlackburn, Michael R.; Vance, Constance O.; Morschl, Eva; Wilson, Constance N.Handbook of Experimental Pharmacology (2009), 193 (Adenosine Receptors in Health and Disease), 215-269CODEN: HEPHD2; ISSN:0171-2004. (Springer GmbH)A review. Extracellular adenosine is produced in a coordinated manner from cells following cellular challenge or tissue injury. Once produced, it serves as an autocrine- and paracrine-signaling mol. through its interactions with seven-membrane-spanning G-protein-coupled adenosine receptors. These signaling pathways have widespread physiol. and pathophysiol. functions. Immune cells express adenosine receptors and respond to adenosine or adenosine agonists in diverse manners. Extensive in vitro and in vivo studies have identified potent anti-inflammatory functions for all of the adenosine receptors on many different inflammatory cells and in various inflammatory disease processes. In addn., specific proinflammatory functions have also been ascribed to adenosine receptor activation. The potent effects of adenosine signaling on the regulation of inflammation suggest that targeting specific adenosine receptor activation or inactivation using selective agonists and antagonists could have important therapeutic implications in numerous diseases. This review is designed to summarize the current status of adenosine receptor signaling in various inflammatory cells and in models of inflammation, with an emphasis on the advancement of adenosine-based therapeutics to treat inflammatory disorders.
- 11Cristalli, G.; Muller, C. E.; Volpini, R. Recent developments in adenosine A2A receptor ligands Handb. Exp. Pharmacol. 2009, 59– 98Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sktr7E&md5=95c3e8fde30c9094d81cea882987ae64Recent developments in adenosine A2A receptor ligandsCristalli, Gloria; Muller, Christa E.; Volpini, RosariaHandbook of Experimental Pharmacology (2009), 193 (Adenosine Receptors in Health and Disease), 59-98CODEN: HEPHD2; ISSN:0171-2004. (Springer GmbH)A review. The development of potent and selective agonists and antagonists of adenosine receptors (ARs) has been a target of medicinal chem. research for several decades, and recently the US Food and Drug Administration has approved Lexiscan, an adenosine deriv. substituted at the 2 position, for use as a pharmacol. stress agent in radionuclide myocardial perfusion imaging. Currently, some other adenosine A2A receptor (A2AAR) agonists and antagonists are undergoing preclin. testing and clin. trials. While agonists are potent antiinflammatory agents also showing hypotensive effects, antagonists are being developed for the treatment of Parkinson's disease. However, since there are still major problems in this field, including side effects, low brain penetration (for the targeting of CNS diseases), short half-life, or lack of in vivo effects, the design and development of new AR ligands is a hot research topic. This review presents an update on the medicinal chem. of A2AAR agonists and antagonists, and stresses the strong need for more selective ligands at the human A2AAR subtype, in particular in the case of agonists.
- 12Poucher, S. M.; Keddie, J. R.; Singh, P.; Stoggall, S. M.; Caulkett, P. W. R.; Jones, G.; Collis, M. G. The in-vitro pharmacology of Zm-241385, a potent, nonxanthine, a(2a) selective adenosine receptor antagonist Br. J. Pharmacol. 1995, 115, 1096– 1102Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXnt1Gksrk%253D&md5=d0f1f44a790e48e5c2c04fe0d71e3638The in vitro pharmacology of ZM 241385, a potent, non-xanthine, A2a selective adenosine receptor antagonistPoucher, S. M.; Keddie, J. R.; Singh, P.; Stoggall, S. M.; Caulkett, P. W. R.; Jones, G.; Collis, M. G.British Journal of Pharmacology (1995), 115 (6), 1096-102CODEN: BJPCBM; ISSN:0007-1188. (Macmillan Scientific & Medical Division)This paper describes the in vitro pharmacol. of ZM 241385 (4-(2-[7-amino-2-(2-furyl) [1,2,4]-triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol), a novel non-xanthine adenosine receptor antagonist with selectivity for the A2 receptor subtype. ZM 241385 had high affinity for A2a receptors. In rat pheochromocytoma cell membranes, ZM 241385 displaced binding of tritiated 5'-N-ethylcarboxamidoadenosine (NECA) with a pIC50 of 9.52, (95% confidence limits, c.l., 9.02-10.02). In guinea-pig isolated Langendorff hearts, ZM 241385 antagonized vasodilation of the coronary bed produced by 2-chloroadenosine (2-CADO) and 2-[p-(2-carboxyethyl) phenylamino]-5'-N-ethylcarboxamidoadenosine (CGS21680) with pA2 values of 8.57 (c.l., 8.45-8.68) and 9.02 (c.l., 8.79-9.24) resp. ZM 241385 had low potency at A2b receptors and antagonized the relaxant effects of adenosine in the guinea-pig aorta with a pA2 of 7.06 (c.l., 6.92-7.19). ZM 241385 had a low affinity at A1 receptors. In rat cerebral cortex membranes it displayed tritiated R-phenylisopropyladenosine (R-PIA) with a pIC50 of 5.69 (c.l., 5.57-5.81). ZM 241385 antagonized the bradycardic action of 2-CADO in guinea-pig atria with a pA2 of 5.95 (c.l., 5.72-6.18). ZM 241385 had low affinity for A3 receptors. At cloned rat A3 receptors expressed in chinese hamster ovary cells, it displayed iodinated aminobenzyl-5'-N-methylcarboxamido adenosine (AB-MECA) with a pIC50 of 3.82 (c.l., 3.67-4.06). ZM 241385 had no significant addnl. pharmacol. effects on the isolated tissues used in these studies at concns. three orders of magnitude greater than those which block A2a receptors. At 10 μM it displayed only minor inhibition of the bradycardic effects in guinea-pig atria to some concns. of carbachol. At 10 μM, ZM 241385 had a small inhibitory effect on relaxant effects of isoprenaline in guinea-pig aorta but no effect on sodium nitrite-induced relaxation. ZM 241385 (100 μM) was without effect on phenylephrine-induced tone in guinea-pig aorta. ZM 241385 (10 μM) had no inhibitory effect on rat hepatocyte phosphodiesterase types I, II, III and IV but caused a small inhibition of the calcium calmodulin-activated type I enzyme. ZM 241385 is the most selective adenosine A2a receptor antagonist yet described and is therefore a useful tool for characterization of responses mediated by A2 adenosine receptors.
- 13Degen, J.; Rarey, M. FlexNovo: structure-based searching in large fragment spaces ChemMedChem 2006, 1, 854– 868Google ScholarThere is no corresponding record for this reference.
- 14Jones, G.; Willett, P.; Glen, R. C.; Leach, A. R.; Taylor, R. Development and validation of a genetic algorithm for flexible docking J. Mol. Biol. 1997, 267, 727– 748Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXis1KntLo%253D&md5=476a2b1d8f80f3ba418052fe29d735caDevelopment and validation of a genetic algorithm for flexible dockingJones, Gareth; Willett, Peter; Glen, Robert C.; Leach, Andrew R.; Taylor, RobinJournal of Molecular Biology (1997), 267 (3), 727-748CODEN: JMOBAK; ISSN:0022-2836. (Academic)Prediction of small mol. binding modes to macromols. of known three-dimensional structure is a problem of paramount importance in rational drug design (the "docking" problem). We report the development and validation of the program GOLD (Genetic Optimization for Ligand Docking). GOLD is an automated ligand docking program that uses a genetic algorithm to explore the full range of ligand conformational flexibility with partial flexibility of the protein and satisfies the fundamental requirement that the ligand must displace loosely bound water on binding. Numerous enhancements and modifications have been applied to the original technique resulting in a substantial increase in the reliability and the applicability of the algorithm. The advanced algorithm has been tested on a dataset of 100 complexes extd. from the Brookhaven Protein Data Bank. When used to dock the ligand back into the binding site, GOLD achieved a 71% success rate in identifying the exptl. binding mode.
- 15Kairys, V.; Fernandes, M. X.; Gilson, M. K. Screening drug-like compounds by docking to homology models: a systematic study J. Chem. Inf. Model. 2006, 46, 365– 379Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht12rtbzL&md5=81e8af8b023309a765cff3e9918a4d4cScreening Drug-Like Compounds by Docking to Homology Models: A Systematic StudyKairys, Visvaldas; Fernandes, Miguel X.; Gilson, Michael K.Journal of Chemical Information and Modeling (2006), 46 (1), 365-379CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In the absence of an exptl. solved structure, a homol. model of a protein target can be used instead for virtual screening of drug candidates by docking and scoring. This approach poses a no. of questions regarding the choice of the template to use in constructing the model, the accuracy of the screening results, and the importance of allowing for protein flexibility. The present study addresses such questions with compd. screening calcns. for multiple homol. models of five drug targets. A central result is that docking to homol. models frequently yields enrichments of known ligands as good as that obtained by docking to a crystal structure of the actual target protein. Interestingly, however, std. measures of the similarity of the template used to build the homol. model to the targeted protein show little correlation with the effectiveness of the screening calcns., and docking to the template itself often is as successful as docking to the corresponding homol. model. Treating key side chains as mobile produces a modest improvement in the results. The reasons for these sometimes unexpected results, and their implications for future methodol. development, are discussed.
- 16Lorber, D. M.; Shoichet, B. K. Flexible ligand docking using conformational ensembles Protein Sci. 1998, 7, 938– 950Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXisFSnurg%253D&md5=64d79dfe6a048c76aec43f8bd9f6bc62Flexible ligand docking using conformational ensemblesLorber, David M.; Shoichet, Brian K.Protein Science (1998), 7 (4), 938-950CODEN: PRCIEI; ISSN:0961-8368. (Cambridge University Press)Mol. docking algorithms suggest possible structures for mol. complexes. They are used to model biol. function and to discover potential ligands. A present challenge for docking algorithms is the treatment of mol. flexibility. Here, the rigid body program, DOCK, is modified to allow it to rapidly fit multiple conformations of ligands. Conformations of a given mol. are pre-calcd. in the same frame of ref., so that each conformer shares a common rigid fragment with all other conformations. The ligand conformers are then docked together, as an ensemble, into a receptor binding site. This takes advantage of the redundancy present in differing conformers of the same mol. The algorithm was tested using three org. ligand protein systems and two protein-protein systems. Both the bound and unbound conformations of the receptors were used. The ligand ensemble method found conformations that resembled those detd. in X-ray crystal structures (RMS values typically less than 1.5 Å). To test the method's usefulness for inhibitor discovery, multi-compd. and multi-conformer databases were screened for compds. known to bind to dihydrofolate reductase and compds. known to bind to thymidylate synthase. In both cases, known inhibitors and substrates were identified in conformations resembling those obsd. exptl. The ligand ensemble method was 100-fold faster than docking a single conformation at a time and was able to screen a database of over 34 million conformations from 117,000 mols. in one to four CPU days on a workstation.
- 17Lorber, D. M.; Shoichet, B. K. Hierarchical docking of databases of multiple ligand conformations Curr. Top. Med. Chem. 2005, 5, 739– 749Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpsFGht74%253D&md5=25c833b5608e5faedc1af3ad38fdfa06Hierarchical docking of databases of multiple ligand conformationsLorber, David M.; Shoichet, Brian K.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2005), 5 (8), 739-749CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)Ligand flexibility is an important problem in mol. docking and virtual screening. To address this challenge, we investigate a hierarchical pre-organization of multiple conformations of small mols. Such organization of pre-calcd. conformations removes the exploration of ligand conformational space from the docking calcn. and allows for concise representation of what can be thousands of conformations. The hierarchy also recognizes and prunes incompatible conformations early in the calcn., eliminating redundant calcns. of fit. We investigate the method by docking the MDL Drug Data Report (MDDR), an annotated database of 100,000 mols., into apo and holo forms of 7 unrelated targets. This annotated database allows us to track the ranking of tens to hundreds of annotated ligands in each of the docking systems. The binding sites and database are prepd. in an automated fashion in an attempt to remove some human bias from the calcns. Many thousands of explicit and implicit ligand conformations may be docked in calcns. not much longer than required for single conformer docking. As long as internal energies are not considered, recombination with the hierarchy is additive as the no. of degrees of freedom is increased. Mols. with even millions of conformations can be docked in a few minutes on a single desktop computer.
- 18Zavodszky, M. I.; Kuhn, L. A. Side-chain flexibility in protein−ligand binding: the minimal rotation hypothesis Protein Sci. 2005, 14, 1104– 1114Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXislOmsrc%253D&md5=1cfcffacb29da4ab665de67b2189dc4eSide-chain flexibility in protein-ligand binding: The minimal rotation hypothesisZavodszky, Maria I.; Kuhn, Leslie A.Protein Science (2005), 14 (4), 1104-1114CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)The goal of this work is to learn from nature about the magnitudes of side-chain motions that occur when proteins bind small org. mols. and model these motions to improve the prediction of protein-ligand complexes. Following anal. of protein side-chain motions upon ligand binding in 63 complexes, we tested the ability of the docking tool SLIDE to model these motions without being restricted to rotameric transitions or deciding which side chains should be considered as flexible. The model tested is that side-chain conformational changes involving more atoms or larger rotations are likely to be more costly and less prevalent than small motions due to energy barriers between rotamers and the potential of large motions to cause new steric clashes. Accordingly, SLIDE adjusts the protein and ligand side groups as little as necessary to achieve steric complementarity. We tested the hypothesis that small motions are sufficient to achieve good dockings using 63 ligands and the apo structures of 20 different proteins and compared SLIDE side-chain rotations to those exptl. obsd. None of these proteins undergoes major main-chain conformational change upon ligand binding, ensuring that side-chain flexibility modeling is not required to compensate for main-chain motions. Although more frugal in the no. of side-chain rotations performed, this model substantially mimics the exptl. obsd. motions. Most side chains do not shift to a new rotamer, and small motions are both necessary and sufficient to predict the correct binding orientation and most protein-ligand interactions for the 20 proteins analyzed.
- 19Kolb, P.; Rosenbaum, D. M.; Irwin, J. J.; Fung, J. J.; Kobilka, B. K.; Shoichet, B. K. Structure-based discovery of beta(2)-adrenergic receptor ligands Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 6843– 6848Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlsV2qsro%253D&md5=4d1a4cb2aa3925aa4c99c6b0496417a7Structure-based discovery of β2-adrenergic receptor ligandsKolb, Peter; Rosenbaum, Daniel M.; Irwin, John J.; Fung, Juan Jose; Kobilka, Brian K.; Shoichet, Brian K.Proceedings of the National Academy of Sciences of the United States of America (2009), 106 (16), 6843-6848CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Aminergic G protein-coupled receptors (GPCRs) have been a major focus of pharmaceutical research for many years. Due partly to the lack of reliable receptor structures, drug discovery efforts have been largely ligand-based. The recently detd. X-ray structure of the β2-adrenergic receptor offers an opportunity to investigate the advantages and limitations inherent in a structure-based approach to ligand discovery against this and related GPCR targets. Approx. 1 million com. available, "lead-like" mols. were docked against the β2-adrenergic receptor structure. On testing of 25 high-ranking mols., 6 were active with binding affinities <4 μM, with the best mol. binding with a Ki of 9 nM (95% confidence interval 7-10 nM). Five of these mols. were inverse agonists. The high hit rate, the high affinity of the most potent mol., the discovery of unprecedented chemotypes among the new inhibitors, and the apparent bias toward inverse agonists among the docking hits, have implications for structure-based approaches against GPCRs that recognize small org. mols.
- 20Sabio, M.; Jones, K.; Topiol, S. Use of the X-ray structure of the beta(2)-adrenergic receptor for drug discovery. Part 2: Identification of active compounds Bioorg. Med. Chem. Lett. 2008, 18, 5391– 5395Google ScholarThere is no corresponding record for this reference.
- 21de Graaf, C.; Rognan, D. Selective structure-based virtual screening for full and partial agonists of the beta 2 adrenergic receptor J. Med. Chem. 2008, 51, 4978– 4985Google ScholarThere is no corresponding record for this reference.
- 22Katritch, V.; Reynolds, K. A.; Cherezov, V.; Hanson, M. A.; Roth, C. B.; Yeager, M.; Abagyan, R. Analysis of full and partial agonists binding to beta(2)-adrenergic receptor suggests a role of transmembrane helix V in agonist-specific conformational changes J. Mol. Recognit. 2009, 22, 307– 318Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXnslGis7k%253D&md5=01e2c841b200697382e551b3cf104451Analysis of full and partial agonists binding to β2-adrenergic receptor suggests a role of transmembrane helix V in agonist-specific conformational changesKatritch, Vsevolod; Reynolds, Kimberly A.; Cherezov, Vadim; Hanson, Michael A.; Roth, Christopher B.; Yeager, Mark; Abagyan, RubenJournal of Molecular Recognition (2009), 22 (4), 307-318CODEN: JMORE4; ISSN:0952-3499. (John Wiley & Sons Ltd.)The 2.4 Å crystal structure of the β2-adrenergic receptor (β2AR) in complex with the high-affinity inverse agonist (-)-carazolol provides a detailed structural framework for the anal. of ligand recognition by adrenergic receptors. Insights into agonist binding and the corresponding conformational changes triggering G-protein coupled receptor (GPCR) activation mechanism are of special interest. While the carazolol pocket captured in the β2AR crystal structure accommodates (-)-isoproterenol and other agonists without steric clashes, a finite movement of the flexible extracellular part of TM-V helix (TM-Ve) obtained by receptor optimization in the presence of docked ligand can further improve the calcd. binding affinities for agonist compds. Tilting of TM-Ve towards the receptor axis provides a more complete description of polar receptor-ligand interactions for full and partial agonists, by enabling optimal engagement of agonists with two exptl. identified anchor sites, formed by Asp 113/Asn 312 and Ser 203/Ser 204/Ser 207 side chains. Further, receptor models incorporating a flexible TM-V backbone allow reliable prediction of binding affinities for a set of diverse ligands, suggesting potential utility of this approach to design of effective and subtype-specific agonists for adrenergic receptors. Systematic differences in capacity of partial, full and inverse agonists to induce TM-V helix tilt in the β2AR model suggest potential role of TM-V as a conformational "rheostat" involved in the whole spectrum of β2AR responses to small mol. signals.
- 23Reynolds, K. A.; Katritch, V.; Abagyan, R. Identifying conformational changes of the beta(2) adrenoceptor that enable accurate prediction of ligand/receptor interactions and screening for GPCR modulators J. Comput.-Aided Mol. Des. 2009, 23, 273– 288Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXkt1yitLw%253D&md5=48e809569368d89fb9ba2c36bddc3f3bIdentifying conformational changes of the β2 adrenoceptor that enable accurate prediction of ligand/receptor interactions and screening for GPCR modulatorsReynolds, Kimberly A.; Katritch, Vsevolod; Abagyan, RubenJournal of Computer-Aided Molecular Design (2009), 23 (5), 273-288CODEN: JCADEQ; ISSN:0920-654X. (Springer)The new β2 Adrenoceptor (β2AR) crystal structures provide a high-resoln. snapshot of receptor interactions with two particular partial inverse agonists, (-)-carazolol and timolol. However, both exptl. and computational studies of GPCR structure are significantly complicated by the existence of multiple conformational states coupled to ligand type and receptor activity. Agonists and antagonists induce or stabilize distinct changes in receptor structure that mediate a range of pharmacol. activities. In this work, the authors (1) established that the existing β2AR crystallog. conformers can be extended to describe ligand/receptor interactions for addnl. antagonist types, (2) generated agonist-bound receptor conformations, and (3) validated these models for agonist and antagonist virtual ligand screening (VLS). Using a ligand directed refinement protocol, the authors derived a single agonist-bound receptor conformation that selectively retrieved a diverse set of full and partial β2AR agonists in VLS trials. Addnl., the impact of extracellular loop two conformation on VLS was assessed by docking studies with rhodopsin-based β2AR homol. models, and loop-deleted receptor models. A general strategy for constructing and selecting agonist-bound receptor pocket conformations is presented, which may prove broadly useful in creating agonist and antagonist bound models for other GPCRs.
- 24Kuntz, I. D.; Blaney, J. M.; Oatley, S. J.; Langridge, R.; Ferrin, T. E. A geometric approach to macromolecule−ligand interactions J. Mol. Biol. 1982, 161, 269– 288Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL38XmtFajsbw%253D&md5=8a4234b24356ea5340f33d906cd71d3eA geometric approach to macromolecule-ligand interactionsKuntz, Irwin D.; Blaney, Jeffrey M.; Oatley, Stuart J.; Langridge, Robert; Ferrin, Thomas E.Journal of Molecular Biology (1982), 161 (2), 269-88CODEN: JMOBAK; ISSN:0022-2836.A method is described to explore geometrically feasible alignments of ligands and receptors of known structure. Algorithms are presented that examine many binding geometries and evaluate them in terms of steric overlap. The procedure uses specific mol. conformations. A method is included for finding putative binding sites on a macromol. surface. Results are reported for heme-myoglobin interaction and the binding of thyroid hormone analogs to prealbumin. In each case, the program finds structures within 1 Å of the x-ray results and also finds distinctly different geometries that provide good steric fits. The approach seems well-suited for generating conformations for energy refinement programs and interactive computer graphics routines.
- 25Shoichet, B. K.; Kuntz, I. D. Matching chemistry and shape in molecular docking Protein Eng. 1993, 6, 723– 732Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXnt1yhtQ%253D%253D&md5=d2ed691493e3dd8701fc19ddd1e7af66Matching chemistry and shape in molecular dockingShoichet, Brian K.; Kuntz, Irwin D.Protein Engineering (1993), 6 (7), 723-32CODEN: PRENE9; ISSN:0269-2139.The authors have added a chem. filter to the ligand placement algorithm of the mol. docking program DOCK. DOCK places ligands in receptors using local shape features. Here the authors label these shape features by chem. type and insist on complementary matches. The authors find fewer phys. unrealistic complexes without reducing the no. of complexes resembling the known ligand-receptor configurations. Approx. 10-fold fewer complexes are calcd. and the new algorithm is correspondingly 10-fold faster than the previous shape-only matching. The authors tested the new algorithm's ability to reproduce three known ligand-receptor complexes: methotrexate in dihydrofolate reductase, deoxyuridine monophosphate in thymidylate synthase and pancreatic trypsin inhibitor in trypsin. The program found configurations within 1 Å of the crystallog. mode, with fewer nonnative solns. compared with shape-only matching. The authors also tested the program's ability to retrieve known inhibitors of thymidylate synthase and dihydrofolate reductase by screening mol. databases against the enzyme structures. Both algorithms retrieved many known inhibitors preferentially to other compds. in the database. The chem. matching algorithm generally ranks known inhibitors better than does matching based on shape alone.
- 26Nicholls, A.; Honig, B. A rapid finite-difference algorithm, utilizing successive over-relaxation to solve the Poisson−Boltzmann equation J. Comput. Chem. 1991, 12, 435– 445Google ScholarThere is no corresponding record for this reference.
- 27Weiner, S. J.; Kollman, P. A.; Case, D. A.; Singh, U. C.; Ghio, C.; Alagona, G.; Profeta, S.; Weiner, P. A new force-field for molecular mechanical simulation of nucleic-acids and proteins J. Am. Chem. Soc. 1984, 106, 765– 784Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXmsVCgug%253D%253D&md5=c4aa27feec7e2c4d34001be10a6cb8e9A new force field for molecular mechanical simulation of nucleic acids and proteinsWeiner, Scott J.; Kollman, Peter A.; Case, David A.; Singh, U. Chandra; Ghio, Caterina; Alagona, Guliano; Profeta, Salvatore, Jr.; Weiner, PaulJournal of the American Chemical Society (1984), 106 (3), 765-84CODEN: JACSAT; ISSN:0002-7863.A force field was developed for simulation of nucleic acids and proteins. The approach began by obtaining equil. bond lengths and angles from microwave, neutron diffraction, and prior mol. mech. calcns., torsional consts. from microwave, NMR, and mol. mech. studies, nonbonded parameters from crystal packing calcns., and at. charges from the fit of a partial charge model to electrostatic potentials calcd. by ab initio quantum mech. theory. The parameters then were refined with mol. mech. studies on the structures and energies of model compds. For nucleic acids, Me Et ether, THF, deoxyadenosine, di-Me phosphate, 9-methylguanine-1-methylcytosine H-bonded complex, 9-methyladenine-1-methylthymine H-bonded complex, and 1,3-dimethyluracil base-stacked dimer were studied. Bond, angle, torsional, nonbonded, and H-bond parameters were varied to optimize the agreement between calcd. and exptl. values for sugar pucker energies and structures, vibrational frequencies of di-Me phosphate and THF, and energies for base pairing and base stacking. For proteins, Φ,ψ maps of glycyl and alanyl dipeptides, H-bonding interactions involving the various protein polar groups, and energy refinement calcns. on insulin were considered. Unlike the models for H bonding involving N and O electron donors, an adequate description of S-H bonding required explicit inclusion of lone pairs.
- 28Babaoglu, K.; Simeonov, A.; Lrwin, J. J.; Nelson, M. E.; Feng, B.; Thomas, C. J.; Cancian, L.; Costi, M. P.; Maltby, D. A.; Jadhav, A.; Inglese, J.; Austin, C. P.; Shoichet, B. K. Comprehensive mechanistic analysis of hits from high-throughput and docking screens against beta-lactamase J. Med. Chem. 2008, 51, 2502– 2511Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXjtFSksb0%253D&md5=7736429aeafc6b17a5d0182e24df0dd4Comprehensive Mechanistic Analysis of Hits from High-Throughput and Docking Screens against β-LactamaseBabaoglu, Kerim; Simeonov, Anton; Irwin, John J.; Nelson, Michael E.; Feng, Brian; Thomas, Craig J.; Cancian, Laura; Costi, M. Paola; Maltby, David A.; Jadhav, Ajit; Inglese, James; Austin, Christopher P.; Shoichet, Brian K.Journal of Medicinal Chemistry (2008), 51 (8), 2502-2511CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)High-throughput screening (HTS) is widely used in drug discovery. Esp. for screens of unbiased libraries, false positives can dominate "hit lists"; their origins are much debated. Here we det. the mechanism of every active hit from a screen of 70,563 unbiased mols. against β-lactamase using quant. HTS (qHTS). Of the 1274 initial inhibitors, 95% were detergent-sensitive and were classified as aggregators. Among the 70 remaining were 25 potent, covalent-acting β-lactams. Mass spectra, counter-screens, and crystallog. identified 12 as promiscuous covalent inhibitors. The remaining 33 were either aggregators or irreproducible. No specific reversible inhibitors were found. We turned to mol. docking to prioritize mols. from the same library for testing at higher concns. Of 16 tested, 2 were modest inhibitors. Subsequent X-ray structures corresponded to the docking prediction. Analog synthesis improved affinity to 8 μM. These results suggest that it may be the phys. behavior of org. mols., not their reactivity, that accounts for most screening artifacts. Structure-based methods may prioritize weak-but-novel chemotypes in unbiased library screens.
- 29Powers, R. A.; Morandi, F.; Shoichet, B. K. Structure-based discovery of a novel, noncovalent inhibitor of AmpC beta-lactamase Structure 2002, 10, 1013– 1023Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XlsVyjsrk%253D&md5=5f539484d08191f0c65626493fc1b262Structure-Based Discovery of a Novel, Noncovalent Inhibitor of AmpC β-LactamasePowers, Rachel A.; Morandi, Federica; Shoichet, Brian K.Structure (Cambridge, MA, United States) (2002), 10 (7), 1013-1023CODEN: STRUE6; ISSN:0969-2126. (Cell Press)β-Lactamases are the most widespread resistance mechanisms to β-lactam antibiotics, and there is a pressing need for novel, non-β-lactam drugs. A database of over 200,000 compds. was docked to the active site of AmpC β-lactamase to identify potential inhibitors. Fifty-six compds. were tested, and three had Ki values of 650 μM or better. The best of these, 3-[(4-chloroanilino)sulfonyl]thiophene-2-carboxylic acid, was a competitive noncovalent inhibitor (Ki = 26 μM), which also reversed resistance to β-lactams in bacteria expressing AmpC. The structure of AmpC in complex with this compd. was detd. by x-ray crystallog. to 1.94 A and reveals that the inhibitor interacts with key active-site residues in sites targeted in the docking calcn. Indeed, the exptl. detd. conformation of the inhibitor closely resembles the prediction. The structure of the enzyme-inhibitor complex presents an opportunity to improve binding affinity in a novel series of inhibitors discovered by structure-based methods.
- 30Meng, E. C.; Shoichet, B. K.; Kuntz, I. D. Automated docking with grid-based energy evaluation J. Comput. Chem. 1992, 13, 505– 524Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38Xit1Omt7c%253D&md5=f472fffa0c9a61652b4c20e4dbbba69eAutomated docking with grid-based energy evaluationMeng, Elaine C.; Shoichet, Brian K.; Kuntz, Irwin D.Journal of Computational Chemistry (1992), 13 (4), 505-24CODEN: JCCHDD; ISSN:0192-8651.The ability to generate feasible binding orientations of a small mol. within a site of known structure is important for ligand design. The authors present a method that combines a rapid, geometric docking algorithm with the evaluation of mol. mechanics interaction energies. The computational costs of evaluation are minimal because the authors precalc. the receptor-dependent terms in the potential function at points on a three-dimensional grid. In four test cases where the components of crystallog. detd. complexes are redocked, the "force field" score correctly identifies the family of orientations closest to the exptl. binding geometry. Scoring functions that consider only steric factors or only electrostatic factors are less successful. The force field function will play an important role in efforts to search databases for potential lead compds.
- 31Shoichet, B. K.; Leach, A. R.; Kuntz, I. D. Ligand solvation in molecular docking Proteins: Struct., Funct., Genet. 1999, 34, 4– 16Google ScholarThere is no corresponding record for this reference.
- 32Wei, B. Q. Q.; Baase, W. A.; Weaver, L. H.; Matthews, B. W.; Shoichet, B. K. A model binding site for testing scoring functions in molecular docking J. Mol. Biol. 2002, 322, 339– 355Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xms1Wqsrs%253D&md5=040e009b33f125ab4abbb322cf32d2c9A Model Binding Site for Testing Scoring Functions in Molecular DockingWei, Binqing Q.; Baase, Walter A.; Weaver, Larry H.; Matthews, Brian W.; Shoichet, Brian K.Journal of Molecular Biology (2002), 322 (2), 339-355CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Science Ltd.)Prediction of interaction energies between ligands and their receptors remains a major challenge for structure-based inhibitor discovery. Much effort has been devoted to developing scoring schemes that can successfully rank the affinities of a diverse set of possible ligands to a binding site for which the structure is known. To test these scoring functions, well-characterized exptl. systems can be very useful. Here, mutation-created binding sites in T4 lysozyme were used to investigate how the quality of at. charges and solvation energies affects mol. docking. At. charges and solvation energies were calcd. for 172,118 mols. in the Available Chems. Directory using a semi-empirical quantum mech. approach by the program AMSOL. The database was first screened against the apolar cavity site created by the mutation Leu99Ala (L99A). Compared to the electronegativity-based charges that are widely used, the new charges and desolvation energies improved ranking of known apolar ligands, and better distinguished them from more polar isosteres that are not obsd. to bind. To investigate whether the new charges had predictive value, the non-polar residue Met102, which forms part of the binding site, was changed to the polar residue glutamine. The structure of the resulting Leu99 Ala and Met102 Gln double mutant of T4 lysozyme (L99A/M102Q) was detd. and the docking calcn. was repeated for the new site. Seven representative polar mols. that preferentially docked to the polar vs. the apolar binding site were tested exptl. All seven bind to the polar cavity (L99A/M102Q) but do not detectably bind to the apolar cavity (L99A). Five ligand-bound structures of L99A/M102Q were detd. by X-ray crystallog. Docking predictions corresponded to the crystallog. results to within 0.4 A RMSD. Improved treatment of partial at. charges and desolvation energies in database docking appears feasible and leads to better distinction of true ligands. Simple model binding sites, such as L99A and its more polar variants, may find broad use in the development and testing of docking algorithms.
- 33Irwin, J. J.; Shoichet, B. K. ZINC—a free database of commercially available compounds for virtual screening J. Chem. Inf. Model. 2005, 45, 177– 182Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVOjt77J&md5=e3892b7dc8608b17a3e63541a5ed60e6ZINC - A Free Database of Commercially Available Compounds for Virtual ScreeningIrwin, John J.; Shoichet, Brian K.Journal of Chemical Information and Computer Sciences (2005), 45 (1), 177-182CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)A crit. barrier to entry into structure-based virtual screening is the lack of a suitable, easy to access database of purchasable compds. We have therefore prepd. a library of 727 842 mols., each with 3D structure, using catalogs of compds. from vendors (the size of this library continues to grow). The mols. have been assigned biol. relevant protonation states and are annotated with properties such as mol. wt., calcd. LogP, and no. of rotatable bonds. Each mol. in the library contains vendor and purchasing information and is ready for docking using a no. of popular docking programs. Within certain limits, the mols. are prepd. in multiple protonation states and multiple tautomeric forms. In one format, multiple conformations are available for the mols. This database is available for free download (http://zinc.docking.org) in several common file formats including SMILES, mol2, 3D SDF, and DOCK flexibase format. A Web-based query tool incorporating a mol. drawing interface enables the database to be searched and browsed and subsets to be created. Users can process their own mols. by uploading them to a server. Our hope is that this database will bring virtual screening libraries to a wide community of structural biologists and medicinal chemists.
- 34Bostrom, J.; Greenwood, J. R.; Gottfries, J. Assessing the performance of OMEGA with respect to retrieving bioactive conformations J. Mol. Graphics Modell. 2003, 21, 449– 462Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXkvFarsQ%253D%253D&md5=6c690a35f8792114a3da7b2afb485140Assessing the performance of OMEGA with respect to retrieving bioactive conformationsBostrom, Jonas; Greenwood, Jeremy R.; Gottfries, JohanJournal of Molecular Graphics & Modelling (2003), 21 (5), 449-462CODEN: JMGMFI; ISSN:1093-3263. (Elsevier Science Inc.)OMEGA is a rule-based program which rapidly generates conformational ensembles of small mols. We have varied the parameters which control the nature of the ensembles generated by OMEGA in a statistical fashion (D-optimal) with the aim of increasing the probability of generating bioactive conformations. Thirty-six drug-like ligands from different ligand-protein complexes detd. by high-resoln. (≤ 2.0 Å) x-ray crystallog. have been analyzed. Statistically significant models (Q2 ≥ 0.75) confirm that one can increase the performance of OMEGA by modifying the parameters. Twenty-eight of the bioactive conformations were retrieved when using a low-energy cut-off (5 kcal/mol), a low RMSD value (0.6 Å) for duplicate removal, and a max. of 1000 output conformations. All of those that were not retrieved had eight or more rotatable bonds. The duplicate removal parameter was found to have the largest impact on retrieval of bioactive conformations, and the max. no. of conformations also affected the results considerably. The input conformation was found to influence the results largely because certain bond angles can prevent the bioactive conformation from being generated as a low-energy conformation. Pre-optimizing the input structures with MMFF94s improved the results significantly. We also investigated the performance of OMEGA in connection with database searching. The shape-matching program Rapid Overlay of Chem. Structures (ROCS) was used as search tool. Two multi-conformational databases were built from the MDDR database plus the 36 compds.; one large (max. 1000 conformations/mol) and one small (max. 100 conformations/mol). Both databases provided satisfactory results in terms of retrieval. ROCS was able to rank 35 out of 36 x-ray structures among the top 500 hits from the large database.
- 35Chambers, C. C.; Hawkins, G. D.; Cramer, C. J.; Truhlar, D. G. Model for aqueous solvation based on class IV atomic charges and first solvation shell effects J. Phys. Chem. 1996, 100, 16385– 16398Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XlsFCqurw%253D&md5=2ff8cd5c0e9d4f37070c0cdf3829fe7eModel for Aqueous Solvation Based on Class IV Atomic Charges and First Solvation Shell EffectsChambers, Candee C.; Cramer, Christopher J.; Truhlar, Donald G.Journal of Physical Chemistry (1996), 100 (40), 16385-16398CODEN: JPCHAX; ISSN:0022-3654. (American Chemical Society)A new set of geometry-based functional forms is presented for parameterizing effective Coulomb radii and at. surface tensions of org. solutes in water. In particular, the radii and surface tensions depend in some cases on distances to nearby atoms. Combining the surface tensions with electrostatic effects included in a Fock operator by the generalized Born model enables one to calc. free energies of solvation, and exptl. free energies of solvation are used to parametrize the theory for water. At. charges are obtained by both the AM1-CM1A and PM3-CM1P class IV charge models, which yield similar results, and hence the same radii and surface tensions are used with both charge models. The authors considered 215 neutral solutes contg. H, C, N, O, F, S, Cl, Br, and I and encompassing a very wide variety of org. functional groups, and a mean unsigned error in the free energy of hydration of 0.50 kcal/mol using CM1A charges and 0.48 kcal/mol using CM1P charges was obtained. The predicted solvation energies for 12 cationic and 22 anionic solutes have mean unsigned deviations from expt. of 4.6 and 4.8 kcal/mol for models based on AM1 and PM3, resp.
- 36Li, J. B.; Zhu, T. H.; Cramer, C. J.; Truhlar, D. G. New class IV charge model for extracting accurate partial charges from wave functions J. Phys. Chem. A 1998, 102, 1820– 1831Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXhtVegtr0%253D&md5=4fe859556188d73500054bb1ebdb0843New Class IV Charge Model for Extracting Accurate Partial Charges from Wave FunctionsLi, Jiabo; Zhu, Tianhai; Cramer, Christopher J.; Truhlar, Donald G.Journal of Physical Chemistry A (1998), 102 (10), 1820-1831CODEN: JPCAFH; ISSN:1089-5639. (American Chemical Society)We propose a new formalism, charge model 2 (CM2), to obtain accurate partial at. charges from a population anal. of wave functions by a parametrized mapping procedure, so that the resulting charges reproduce highly accurate charge-dependent observables. The new method, which produces class IV charges, is illustrated by developing CM2 mappings of Lowdin charges obtained from semiempirical and ab initio Hartree-Fock theory and d. functional theory, in particular AM1, PM3, HF/MIDI!, HF/6-31G*, HF/6-31+G*, BPW91/MIDI!, BPW91/6-31G*, B3LYP/MIDI!, and BPW91/DZVP calcns. The CM2 partial charges reproduce exptl. dipole moments with root-mean-square errors that are typically a factor of 7 better than dipole moments computed from Mulliken population anal., a factor of 3 better than dipole moments computed by Lowdin anal., and even a factor of 2 better than dipole moments computed from the continuous electron d. At the HF/6-31G* and B3LYP/MIDI! levels, the new charge model yields root-mean-square errors of 0.19 and 0.18 D, resp., for the dipole moments of a set of 211 polar mols. contg. a diverse range of structures and org. functional groups and the elements H, C, N, O, F, Si, P, S, Cl, Br, and I. A comparison shows that the new charge model predicts dipole moments more accurately than MP2/cc-pVDZ calcns., which are considerably more expensive. The quality of the results is similarly good for electrostatic potentials and for the other parametrizations as well.
- 37Weiner, S. J.; Kollman, P. A.; Nguyen, D. T.; Case, D. A. An all atom force-field for simulations of proteins and nucleic-acids J. Comput. Chem. 1986, 7, 230– 252Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XhvVarsLY%253D&md5=33f0620f30d45081f9383240f4dacd73An all atom force field for simulations of proteins and nucleic acidsWeiner, Scott J.; Kollman, Peter A.; Nguyen, Dzung T.; Case, David A.Journal of Computational Chemistry (1986), 7 (2), 230-52CODEN: JCCHDD; ISSN:0192-8651.An all-atom potential energy function for the simulation of proteins and nucleic acids is presented. This work is an extension of the CH united atom function recently presented by S. J. Weiner et al. (1984). The parameters of this function are based on calcns. on ethane, propane, n-butane, di-Me ether, Me Et ether, THF, imidazole, indole, deoxyadenosine, base-paired dinucleoside phosphates, adenine, guanine, uracil, cytosine, thymine, insulin, and myoglobin. These parameters were also used to carry out the 1st general vibrational anal. of all 5 nucleic acid bases with a mol. mechanics potential approach.
- 38http://accelrys.com/products/scitegic/.Google ScholarThere is no corresponding record for this reference.
- 39Olah, M.; Mracec, M.; Ostopovici, L.; Rad, R.; Bora, A.; Hadaruga, N.; Olah, I.; Banda, M.; Simon, Z.; Mracec, M.; Oprea, T. I. WOMBAT: World of Molecular Bioactivity. In Chemoinformatics in Drug Discovery; Oprea, T. I., Ed.; Wiley-VCH: Weinheim, Germany, 2005; pp 221− 239.Google ScholarThere is no corresponding record for this reference.
- 40http://www.ebi.ac.uk/chembl.Google ScholarThere is no corresponding record for this reference.
- 41Keiser, M. J.; Roth, B. L.; Armbruster, B. N.; Ernsberger, P.; Irwin, J. J.; Shoichet, B. K. Relating protein pharmacology by ligand chemistry Nat. Biotechnol. 2007, 25, 197– 206Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlOrsLo%253D&md5=1b7373d52563fca5fe1e893d85f70573Relating protein pharmacology by ligand chemistryKeiser, Michael J.; Roth, Bryan L.; Armbruster, Blaine N.; Ernsberger, Paul; Irwin, John J.; Shoichet, Brian K.Nature Biotechnology (2007), 25 (2), 197-206CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The identification of protein function based on biol. information is an area of intense research. Here the authors consider a complementary technique that quant. groups and relates proteins based on the chem. similarity of their ligands. The authors began with 65,000 ligands annotated into sets for hundreds of drug targets. The similarity score between each set was calcd. using ligand topol. A statistical model was developed to rank the significance of the resulting similarity scores, which are expressed as a min. spanning tree to map the sets together. Although these maps are connected solely by chem. similarity, biol. sensible clusters nevertheless emerged. Links among unexpected targets also emerged, among them that methadone, emetine and loperamide (Imodium) may antagonize muscarinic M3, α2 adrenergic and neurokinin NK2 receptors, resp. These predictions were subsequently confirmed exptl. Relating receptors by ligand chem. organizes biol. to reveal unexpected relationships that may be assayed using the ligands themselves.
- 42Tondi, D.; Morandi, F.; Bonnet, R.; Costi, M. P.; Shoichet, B. K. Structure-based optimization of a non-beta-lactam lead results in inhibitors that do not up-regulate beta-lactamase expression in cell culture J. Am. Chem. Soc. 2005, 127, 4632– 4639Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXit1Krsbk%253D&md5=ddad35ac3e6fb96cf65e12cd807f271cStructure-Based Optimization of a Non-β-lactam Lead Results in Inhibitors That Do Not Up-Regulate β-Lactamase Expression in Cell CultureTondi, Donatella; Morandi, Federica; Bonnet, Richard; Costi, M. Paola; Shoichet, Brian K.Journal of the American Chemical Society (2005), 127 (13), 4632-4639CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Bacterial expression of β-lactamases is the most widespread resistance mechanism to β-lactam antibiotics, such as penicillins and cephalosporins. There is a pressing need for novel, non-β-lactam inhibitors of these enzymes. One previously discovered novel inhibitor of the β-lactamase AmpC (I) has several favorable properties: it is chem. dissimilar to β-lactams and is a noncovalent, competitive inhibitor of the enzyme. However, at 26 μM its activity is modest. Using the x-ray structure of the AmpC/I complex as a template, 14 analogs were designed and synthesized. The most active of these, (II), had a Ki of 1 μM, 26-fold better than the lead. To understand the origins of this improved activity, the structures of AmpC in complex with compd. II and an analog were detd. by x-ray crystallog. to 1.97 and 1.96 Å, resp. II was active in cell culture, reversing resistance to the third generation cephalosporin ceftazidime in bacterial pathogens expressing AmpC. In contrast to β-lactam-based inhibitors clavulanate and cefoxitin, compd. II did not up-regulate β-lactamase expression in cell culture but simply inhibited the enzyme expressed by the resistant bacteria. Its escape from this resistance mechanism derives from its dissimilarity to β-lactam antibiotics.
- 43Jarvis, M. F.; Schulz, R.; Hutchison, A. J.; Do, U. H.; Sills, M. A.; Williams, M. [H-3] Cgs-21680, a selective A2 adenosine receptor agonist directly labels A2-receptors in rat-brain J. Pharmacol. Exp. Ther. 1989, 251, 888– 893Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXhtFamur4%253D&md5=fc1961da1f86a5592fa6142878952fa9[3H]CGS 21680, a selective A2 adenosine receptor agonist directly labels A2 receptors in rat brainJarvis, Michael F.; Schulz, Rainer; Hutchison, Alan J.; Do, Un Hoi; Sills, Matthew A.; Williams, MichaelJournal of Pharmacology and Experimental Therapeutics (1989), 251 (3), 888-93CODEN: JPETAB; ISSN:0022-3565.Characterization of the adenosine A2 receptor has been limited due to the lack of available ligands which have high affinity and selectivity for this adenosine receptor subtype. In the present study, the binding of a highly A2-selective agonist radioligand, [3H]CGS 21680 (I) is described. [3H]CGS 21680 specific binding to rat striatal membranes was saturable, reversible, and dependent on protein concn. Satn. studies revealed that [3H]CGS 21680 bound with high affinity (Kd =15.5 nM) and limited capacity (apparent Bmax = 375 fmol/mg protein) to a single class of recognition sites. Ests. of ligand affinity (16 nM) detd. from assocn. and dissocn. kinetic expts. were in close agreement with the results from the satn. studies. [3H]CGS 21680 binding was greatest in striatal membranes with negligible specific binding obtained in rat cortical membranes. Adenosine agonists ligands competed for the binding of 5 nM [3H]CGS 21680 to striatal membranes with the following order of activity; CGS 21680 = 5'-N-ethylcarboxamidoadenosine > 2-phenylaminoadenosine (CV-1808) ; 5'-N-methylcarboxamidoadenosine = 2-chloroadenosine > R-phenylisopropyladenosine > N6-cyclohexyladenosine > N6-cyclopentyltheophylline > S-phenylisopropyladenosine. The nonxanthine adenosine antagonist, CGS 15943A, was the most active compd. in inhibiting the binding of [3H]CGS 21680. Other adenosine antagonists inhibited binding in the following order; xanthine amine congener = (1,3-dipropyl-8-(2-amino-4-chloro)phenylxanthine > 1,3-dipropyl-8-cyclopentylxanthine > 1,3-diethyl-8-phenylxanthine > 8-phenyltheophylline > 8-cyclopentyltheophylline = xanthine carboxylic acid congener > 8-parasulfophenyltheophylline > theophylline > caffeine. The pharmacol. profile of both adenosine agonist and antagonist compds. to compete for the binding of [3H]CGS 21680 was consistent with a selective interaction at the high affinity adenosine A2 receptor. A high pos. correlation was obsd. between the pharmacol. profile of adenosine ligands to inhibit the binding of [3H]CGS 21680 and the selective binding of [3H]NECA (+50 nM CPA) to high affinity A2 receptors. However, some differences between these assays were found for compds. which have moderate affinity and nonselective actions at both the A1 and A2 adenosine receptor subtypes. Unlike data obtained with nonselective adenosine ligands, the present results indicate that [3H]CGS 21680 directly labels the high affinity A2 receptor in rat brain without the need to block binding activity at the A1 receptor. The high degree of selectivity (>170-fold) and high affinity of [3H]CGS 21680 make this the current ligand of choice for the in vitro characterization of high affinity A2 receptors.
- 44Klotz, K. N.; Lohse, M. J.; Schwabe, U.; Cristalli, G.; Vittori, S.; Grifantini, M. 2-Chloro-N-6-[H-3]cyclopentyladenosine ([H-3]Ccpa), a high-affinity agonist radioligand for A1 adenosine receptors Naunyn-Schmiedeberg's Arch. Pharmacol. 1989, 340, 679– 683Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXhtFGhsL8%253D&md5=d44acdedb7aa0c3b3c6f7d79666ae1392-Chloro-N6-[3H]cyclopentyladenosine ([3H]CPPA) - a high affinity agonist radioligand for A1 adenosine receptorsKlotz, Karl Norbert; Lohse, Martin J.; Schwabe, Ulrich; Cristalli, Gloria; Vittori, Sauro; Grifantini, MarioNaunyn-Schmiedeberg's Archives of Pharmacology (1989), 340 (6), 679-83CODEN: NSAPCC; ISSN:0028-1298.The tritiated analog of 2-chloro-N6-cyclopentyladenosine (CCPA) (I), an adenosine deriv. with subnanomolar affinity and a 10,000-fold selectivity for A1 adenosine receptors, has been examd. as a new agonist radioligand. [3H]CCPA was prepd. with a specific radioactivity of 1.58 TBq/mmol (43 Ci/mmol) and bound in a reversible manner to A1 receptors from rat brain membranes with a high affinity KD value of 0.2 nmol/L. In the presence of GTP, a KDvalue of 13 nmol/L was detd. for the low affinity state for agonist binding. Competition of several adenosine receptor agonists and antagonists for [3H]CCPA binding to rat brain membranes confirmed binding to an A1 receptor. Solubilized A1 receptors bound [3H]CCPA with similar affinity for the high affinity state. At solubilized receptors a reduced assocn. rate was obsd. in the presence of MgCl2, as has been shown for the agonist [3H]N6-phenylisopropyladenosine ([3H]PIA). [3H]CCPA was also used for detection of A1 receptors in rat cardiomyocyte membranes, a tissue with a very low receptor d. Kd-Value of 0.4 nmol-L and a Bmax-value of 16 fmol-platelet membranes, no specific binding of [3H]CCPA was measured at concns. up to 400 nmol/L, indicating that A2 receptors did not bind [3H]CCPA. Based on the subnanomolar affinity and the high selectivity for A1 receptors, [3H]CCPA proved to be a useful agonist radioligand for characterization of A1 adenosine receptors also in tissues with very low receptor d.
- 45Olah, M. E.; Gallorodriguez, C.; Jacobson, K. A.; Stiles, G. L. I-125 4-aminobenzyl-5′-N-methylcarboxamidoadenosine, a high-affinity radioligand for the rat a(3) adenosine receptor Mol. Pharmacol. 1994, 45, 978– 982Google ScholarThere is no corresponding record for this reference.
- 46Englert, M.; Quitterer, U.; Klotz, K. N. Effector coupling of stably transfected human A(3) adenosine receptors in CHO cells Biochem. Pharmacol. 2002, 64, 61– 65Google ScholarThere is no corresponding record for this reference.
- 47Jacobson, K. A.; Park, K. S.; Jiang, J. L.; Kim, Y. C.; Olah, M. E.; Stiles, G. L.; Ji, X. D. Pharmacological characterization of novel A(3) adenosine receptor-selective antagonists Neuropharmacology 1997, 36, 1157– 1165Google ScholarThere is no corresponding record for this reference.
- 48Nordstedt, C.; Fredholm, B. B. A modification of a protein-binding method for rapid quantification of camp in cell-culture supernatants and body-fluid Anal. Biochem. 1990, 189, 231– 234Google ScholarThere is no corresponding record for this reference.
- 49Post, S. R.; Ostrom, R. S.; Insel, P. A. Biochemical methods for detection and measurement of cyclic AMP and adenylyl cyclase activity Methods Mol. Biol. 2000, 126, 363– 374Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXjslWmuw%253D%253D&md5=0c9a58e800650d2f7d0ea512017472a0Biochemical methods for detection and measurement of cyclic AMP and adenylyl cyclase activityPost, Steven R.; Ostrom, Rennolds S.; Insel, Paul A.Methods in Molecular Biology (Totowa, New Jersey) (2000), 126 (Adrenergic Receptor Protocols), 363-374CODEN: MMBIED; ISSN:1064-3745. (Humana Press Inc.)Several assays are described and detailed for the characterization and anal. of cAMP and adenylyl cyclase.
- 50Bradford, M. M. Rapid and sensitive method for quantitation of microgram quantities of protein utilizing principle of protein−dye binding Anal. Biochem. 1976, 72, 248– 254Google ScholarThere is no corresponding record for this reference.
- 51McGovern, S. L.; Helfand, B. T.; Feng, B.; Shoichet, B. K. A specific mechanism of nonspecific inhibition J. Med. Chem. 2003, 46, 4265– 4272Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXmsFamsbs%253D&md5=cd0fd6bf0a64ceb7d7219dae2406dd21A Specific Mechanism of Nonspecific InhibitionMcGovern, Susan L.; Helfand, Brian T.; Feng, Brian; Shoichet, Brian K.Journal of Medicinal Chemistry (2003), 46 (20), 4265-4272CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Promiscuous small mols. plague screening libraries and hit lists. Previous work has found that several nonspecific compds. form submicrometer aggregates, and it has been suggested that this aggregate species is responsible for the inhibition of many different enzymes. It is not understood how aggregates inhibit their targets. To address this question, biophys., kinetic, and microscopy methods were used to study the interaction of promiscuous, aggregate-forming inhibitors with model proteins. By use of centrifugation and gel electrophoresis, aggregates and protein were found to directly interact. This is consistent with a subsequent observation from confocal fluorescence microscopy that aggregates conc. green fluorescent protein. β-Lactamase mutants with increased or decreased thermodn. stability relative to wild-type enzyme were equally inhibited by an aggregate-forming compd., suggesting that denaturation by unfolding was not the primary mechanism of interaction. Instead, visualization by electron microscopy revealed that enzyme assocs. with the surface of inhibitor aggregates. This assocn. could be reversed or prevented by the addn. of Triton X-100. These observations suggest that the aggregates formed by promiscuous compds. reversibly sequester enzyme, resulting in apparent inhibition. They also suggest a simple method to identify or reverse the action of aggregate-based inhibitors, which appear to be widespread.
- 52Kim, J. H.; Wess, J.; Vanrhee, A. M.; Schoneberg, T.; Jacobson, K. A. Site-directed mutagenesis identifies residues involved in ligand recognition in the human a(2a) adenosine receptor J. Biol. Chem. 1995, 270, 13987– 13997Google ScholarThere is no corresponding record for this reference.
- 53Webb, T. R.; Lvovskiy, D.; Kim, S. A.; Ji, X. D.; Melman, N.; Linden, J.; Jacobson, K. A. Quinazolines as adenosine receptor antagonists: SAR and selectivity for A(2B) receptors Bioorg. Med. Chem. 2003, 11, 77– 85Google ScholarThere is no corresponding record for this reference.
- 54Hert, J.; Irwin, J. J.; Laggner, C.; Keiser, M. J.; Shoichet, B. K. Quantifying biogenic bias in screening libraries Nat. Chem. Biol. 2009, 5, 479– 483Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXms1aksbg%253D&md5=67a631cb210e4cf7aa56190f7e2d7e2bQuantifying biogenic bias in screening librariesHert, Jerome; Irwin, John J.; Laggner, Christian; Keiser, Michael J.; Shoichet, Brian K.Nature Chemical Biology (2009), 5 (7), 479-483CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)In lead discovery, libraries of 106 mols. are screened for biol. activity. Given the over 1060 drug-like mols. thought possible, such screens might never succeed. The fact that they do, even occasionally, implies a biased selection of library mols. We have developed a method to quantify the bias in screening libraries toward biogenic mols. With this approach, we consider what is missing from screening libraries and how they can be optimized.
- 55Soelaiman, S.; Wei, B. Q.; Bergson, P.; Lee, Y. S.; Shen, Y.; Mrksich, M.; Shoichet, B. K.; Tang, W. J. Structure-based inhibitor discovery against adenylyl cyclase toxins from pathogenic bacteria that cause anthrax and whooping cough J. Biol. Chem. 2003, 278, 25990– 25997Google ScholarThere is no corresponding record for this reference.
- 56Katritch, V.; Jaakola, V. P.; Lane, J. R.; Lin, J.; Ijzerman, A. P.; Yeager, M.; Kufareva, I.; Stevens, R. C.; Abagyan, R. Structure-based discovery of novel chemotypes for adenosine A(2A) receptor antagonists J. Med. Chem. 2010, 53, 1799– 1809Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptlKqsw%253D%253D&md5=5a54df43f6edd20d83e7e5942e2f9811Structure-Based Discovery of Novel Chemotypes for Adenosine A2A Receptor AntagonistsKatritch, Vsevolod; Jaakola, Veli-Pekka; Lane, J. Robert; Lin, Judy; IJzerman, Adriaan P.; Yeager, Mark; Kufareva, Irina; Stevens, Raymond C.; Abagyan, RubenJournal of Medicinal Chemistry (2010), 53 (4), 1799-1809CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent progress in crystallog. of G-protein coupled receptors opens an unprecedented venue for structure-based GPCR drug discovery. To test efficiency of the structure-based approach, we performed mol. docking and virtual ligand screening (VLS) of more than 4 million com. available "drug-like" and "lead-like" compds. against the A2AAR 2.6 Å resoln. crystal structure. Out of 56 high ranking compds. tested in A2AAR binding assays, 23 showed affinities under 10 μM, 11 of those had sub-μM affinities and two compds. had affinities under 60 nM. The identified hits represent at least 9 different chem. scaffolds and are characterized by very high ligand efficiency (0.3-0.5 kcal/mol per heavy atom). Significant A2AAR antagonist activities were confirmed for 10 out of 13 ligands tested in functional assays. High success rate, novelty, and diversity of the chem. scaffolds and strong ligand efficiency of the A2AAR antagonists identified in this study suggest practical applicability of receptor-based VLS in GPCR drug discovery.
Cited By
This article is cited by 191 publications.
- Hyeongwoo Kim, Kyunghoon Lee, Chansu Kim, Jaechang Lim, Woo Youn Kim. DFRscore: Deep Learning-Based Scoring of Synthetic Complexity with Drug-Focused Retrosynthetic Analysis for High-Throughput Virtual Screening. Journal of Chemical Information and Modeling 2023, Article ASAP.
- Margarita Stampelou, Anna Suchankova, Efpraxia Tzortzini, Lakshiv Dhingra, Kerry Barkan, Nikolaos Lougiakis, Panagiotis Marakos, Nicole Pouli, Graham Ladds, Antonios Kolocouris. Dual A1/A3 Adenosine Receptor Antagonists: Binding Kinetics and Structure−Activity Relationship Studies Using Mutagenesis and Alchemical Binding Free Energy Calculations. Journal of Medicinal Chemistry 2022, 65
(19)
, 13305-13327. https://doi.org/10.1021/acs.jmedchem.2c01123
- Vigneshwaran Namasivayam, Katja Silbermann, Michael Wiese, Jens Pahnke, Sven Marcel Stefan. C@PA: Computer-Aided Pattern Analysis to Predict Multitarget ABC Transporter Inhibitors. Journal of Medicinal Chemistry 2021, 64
(6)
, 3350-3366. https://doi.org/10.1021/acs.jmedchem.0c02199
- Celeste N. Alverez, Jung-Eun Park, Kiran S. Toti, Yangliu Xia, Kristopher W. Krausz, Ganesha Rai, Jeong K. Bang, Frank J. Gonzalez, Kenneth A. Jacobson, Kyung S. Lee. Identification of a New Heterocyclic Scaffold for Inhibitors of the Polo-Box Domain of Polo-like Kinase 1. Journal of Medicinal Chemistry 2020, 63
(22)
, 14087-14117. https://doi.org/10.1021/acs.jmedchem.0c01669
- Eleonora Comeo, Nicholas D. Kindon, Mark Soave, Leigh A. Stoddart, Laura E. Kilpatrick, Peter J. Scammells, Stephen J. Hill, Barrie Kellam. Subtype-Selective Fluorescent Ligands as Pharmacological Research Tools for the Human Adenosine A2A Receptor. Journal of Medicinal Chemistry 2020, 63
(5)
, 2656-2672. https://doi.org/10.1021/acs.jmedchem.9b01856
- Flavio Ballante, Axel Rudling, Alexey Zeifman, Andreas Luttens, Duy Duc Vo, John J. Irwin, Jan Kihlberg, Jose Brea, Maria Isabel Loza, Jens Carlsson. Docking Finds GPCR Ligands in Dark Chemical Matter. Journal of Medicinal Chemistry 2020, 63
(2)
, 613-620. https://doi.org/10.1021/acs.jmedchem.9b01560
- Panagiotis Lagarias, Kerry Barkan, Eva Tzortzini, Margarita Stampelou, Eleni Vrontaki, Graham Ladds, Antonios Kolocouris. Insights to the Binding of a Selective Adenosine A3 Receptor Antagonist Using Molecular Dynamic Simulations, MM-PBSA and MM-GBSA Free Energy Calculations, and Mutagenesis. Journal of Chemical Information and Modeling 2019, 59
(12)
, 5183-5197. https://doi.org/10.1021/acs.jcim.9b00751
- Dahlia
R. Weiss, Joel Karpiak, Xi-Ping Huang, Maria F. Sassano, Jiankun Lyu, Bryan L. Roth, Brian K. Shoichet. Selectivity Challenges in Docking Screens for GPCR Targets and Antitargets. Journal of Medicinal Chemistry 2018, 61
(15)
, 6830-6845. https://doi.org/10.1021/acs.jmedchem.8b00718
- Mariama Jaiteh, Alexey Zeifman, Marcus Saarinen, Per Svenningsson, Jose Bréa, Maria Isabel Loza, Jens Carlsson. Docking Screens for Dual Inhibitors of Disparate Drug Targets for Parkinson’s Disease. Journal of Medicinal Chemistry 2018, 61
(12)
, 5269-5278. https://doi.org/10.1021/acs.jmedchem.8b00204
- Panagiotis Lagarias, Eleni Vrontaki, George Lambrinidis, Dimitrios Stamatis, Marino Convertino, Gabriella Ortore, Thomas Mavromoustakos, Karl-Norbert Klotz, Antonios Kolocouris. Discovery of Novel Adenosine Receptor Antagonists through a Combined Structure- and Ligand-Based Approach Followed by Molecular Dynamics Investigation of Ligand Binding Mode. Journal of Chemical Information and Modeling 2018, 58
(4)
, 794-815. https://doi.org/10.1021/acs.jcim.7b00455
- Yoonji Lee, Shaherin Basith, and Sun Choi . Recent Advances in Structure-Based Drug Design Targeting Class A G Protein-Coupled Receptors Utilizing Crystal Structures and Computational Simulations. Journal of Medicinal Chemistry 2018, 61
(1)
, 1-46. https://doi.org/10.1021/acs.jmedchem.6b01453
- Inbar Fish, Anne Stößel, Katrin Eitel, Celine Valant, Sabine Albold, Harald Huebner, Dorothee Möller, Mary J. Clark, Roger K. Sunahara, Arthur Christopoulos, Brian K. Shoichet, and Peter Gmeiner . Structure-Based Design and Discovery of New M2 Receptor Agonists. Journal of Medicinal Chemistry 2017, 60
(22)
, 9239-9250. https://doi.org/10.1021/acs.jmedchem.7b01113
- Sheng Tian, Xu Wang, Linlang Li, Xiaohu Zhang, Youyong Li, Feng Zhu, Tingjun Hou, and Xuechu Zhen . Discovery of Novel and Selective Adenosine A2A Receptor Antagonists for Treating Parkinson’s Disease through Comparative Structure-Based Virtual Screening. Journal of Chemical Information and Modeling 2017, 57
(6)
, 1474-1487. https://doi.org/10.1021/acs.jcim.7b00188
- Marta Arimont, Shan-Liang Sun, Rob Leurs, Martine Smit, Iwan J. P. de Esch, and Chris de Graaf . Structural Analysis of Chemokine Receptor–Ligand Interactions. Journal of Medicinal Chemistry 2017, 60
(12)
, 4735-4779. https://doi.org/10.1021/acs.jmedchem.6b01309
- Zhong Zheng, Xi-Ping Huang, Thomas J. Mangano, Rodger Zou, Xin Chen, Saheem A. Zaidi, Bryan L. Roth, Raymond C. Stevens, and Vsevolod Katritch . Structure-Based Discovery of New Antagonist and Biased Agonist Chemotypes for the Kappa Opioid Receptor. Journal of Medicinal Chemistry 2017, 60
(7)
, 3070-3081. https://doi.org/10.1021/acs.jmedchem.7b00109
- Anirudh Ranganathan, Philipp Heine, Axel Rudling, Andreas Plückthun, Lutz Kummer, and Jens Carlsson . Ligand Discovery for a Peptide-Binding GPCR by Structure-Based Screening of Fragment- and Lead-Like Chemical Libraries. ACS Chemical Biology 2017, 12
(3)
, 735-745. https://doi.org/10.1021/acschembio.6b00646
- Thomas M. Frimurer, Franziska Mende, Anne-Sofie Graae, Maja S. Engelstoft, Kristoffer L. Egerod, Rie Nygaard, Lars-Ole Gerlach, Jakob Bondo Hansen, Thue W. Schwartz, and Birgitte Holst . Model-Based Discovery of Synthetic Agonists for the Zn2+-Sensing G-Protein-Coupled Receptor 39 (GPR39) Reveals Novel Biological Functions. Journal of Medicinal Chemistry 2017, 60
(3)
, 886-898. https://doi.org/10.1021/acs.jmedchem.6b00648
- Ali Jazayeri, Stephen P. Andrews, and Fiona H. Marshall . Structurally Enabled Discovery of Adenosine A2A Receptor Antagonists. Chemical Reviews 2017, 117
(1)
, 21-37. https://doi.org/10.1021/acs.chemrev.6b00119
- David Rodríguez, Saibal Chakraborty, Eugene Warnick, Steven Crane, Zhan-Guo Gao, Robert O’Connor, Kenneth A. Jacobson, and Jens Carlsson . Structure-Based Screening of Uncharted Chemical Space for Atypical Adenosine Receptor Agonists. ACS Chemical Biology 2016, 11
(10)
, 2763-2772. https://doi.org/10.1021/acschembio.6b00357
- Ainoleena Turku, Alexandre Borrel, Teppo O. Leino, Lasse Karhu, Jyrki P. Kukkonen, and Henri Xhaard . Pharmacophore Model To Discover OX1 and OX2 Orexin Receptor Ligands. Journal of Medicinal Chemistry 2016, 59
(18)
, 8263-8275. https://doi.org/10.1021/acs.jmedchem.6b00333
- John J. Irwin and Brian K. Shoichet . Docking Screens for Novel Ligands Conferring New Biology. Journal of Medicinal Chemistry 2016, 59
(9)
, 4103-4120. https://doi.org/10.1021/acs.jmedchem.5b02008
- Yu Zhou, Jing Ma, Xingyu Lin, Xi-Ping Huang, Kaichun Wu, and Niu Huang . Structure-Based Discovery of Novel and Selective 5-Hydroxytryptamine 2B Receptor Antagonists for the Treatment of Irritable Bowel Syndrome. Journal of Medicinal Chemistry 2016, 59
(2)
, 707-720. https://doi.org/10.1021/acs.jmedchem.5b01631
- Anirudh Ranganathan, Leigh A. Stoddart, Stephen J. Hill, and Jens Carlsson . Fragment-Based Discovery of Subtype-Selective Adenosine Receptor Ligands from Homology Models. Journal of Medicinal Chemistry 2015, 58
(24)
, 9578-9590. https://doi.org/10.1021/acs.jmedchem.5b01120
- Teague Sterling and John J. Irwin . ZINC 15 – Ligand Discovery for Everyone. Journal of Chemical Information and Modeling 2015, 55
(11)
, 2324-2337. https://doi.org/10.1021/acs.jcim.5b00559
- Albert J. Kooistra, Rob Leurs, Iwan J. P. de Esch, and Chris de Graaf . Structure-Based Prediction of G-Protein-Coupled Receptor Ligand Function: A β-Adrenoceptor Case Study. Journal of Chemical Information and Modeling 2015, 55
(5)
, 1045-1061. https://doi.org/10.1021/acs.jcim.5b00066
- David Rodríguez, Zhang-Guo Gao, Steven M. Moss, Kenneth A. Jacobson, and Jens Carlsson . Molecular Docking Screening Using Agonist-Bound GPCR Structures: Probing the A2A Adenosine Receptor. Journal of Chemical Information and Modeling 2015, 55
(3)
, 550-563. https://doi.org/10.1021/ci500639g
- Andrew Anighoro, Dagmar Stumpfe, Kathrin Heikamp, Kristin Beebe, Leonard M. Neckers, Jürgen Bajorath, and Giulio Rastelli . Computational Polypharmacology Analysis of the Heat Shock Protein 90 Interactome. Journal of Chemical Information and Modeling 2015, 55
(3)
, 676-686. https://doi.org/10.1021/ci5006959
- Denis Schmidt, Viachaslau Bernat, Regine Brox, Nuska Tschammer, and Peter Kolb . Identifying Modulators of CXC Receptors 3 and 4 with Tailored Selectivity Using Multi-Target Docking. ACS Chemical Biology 2015, 10
(3)
, 715-724. https://doi.org/10.1021/cb500577j
- Dahlia A. Goldfeld, Robert Murphy, Byungchan Kim, Lingle Wang, Thijs Beuming, Robert Abel, and Richard A. Friesner . Docking and Free Energy Perturbation Studies of Ligand Binding in the Kappa Opioid Receptor. The Journal of Physical Chemistry B 2015, 119
(3)
, 824-835. https://doi.org/10.1021/jp5053612
- Andrew Anighoro, Jürgen Bajorath, and Giulio Rastelli . Polypharmacology: Challenges and Opportunities in Drug Discovery. Journal of Medicinal Chemistry 2014, 57
(19)
, 7874-7887. https://doi.org/10.1021/jm5006463
- Antonella Ciancetta, Alberto Cuzzolin, and Stefano Moro . Alternative Quality Assessment Strategy to Compare Performances of GPCR-Ligand Docking Protocols: The Human Adenosine A2A Receptor as a Case Study. Journal of Chemical Information and Modeling 2014, 54
(8)
, 2243-2254. https://doi.org/10.1021/ci5002857
- David Rodríguez, Anirudh Ranganathan, and Jens Carlsson . Strategies for Improved Modeling of GPCR-Drug Complexes: Blind Predictions of Serotonin Receptors Bound to Ergotamine. Journal of Chemical Information and Modeling 2014, 54
(7)
, 2004-2021. https://doi.org/10.1021/ci5002235
- Eelke B. Lenselink, Thijs Beuming, Woody Sherman, Herman W. T. van Vlijmen, and Adriaan P. IJzerman . Selecting an Optimal Number of Binding Site Waters To Improve Virtual Screening Enrichments Against the Adenosine A2A Receptor. Journal of Chemical Information and Modeling 2014, 54
(6)
, 1737-1746. https://doi.org/10.1021/ci5000455
- Dan Chen, Anirudh Ranganathan, Adriaan P. IJzerman, Gregg Siegal, and Jens Carlsson . Complementarity between in Silico and Biophysical Screening Approaches in Fragment-Based Lead Discovery against the A2A Adenosine Receptor. Journal of Chemical Information and Modeling 2013, 53
(10)
, 2701-2714. https://doi.org/10.1021/ci4003156
- Magdalena Bacilieri, Antonella Ciancetta, Silvia Paoletta, Stephanie Federico, Sandro Cosconati, Barbara Cacciari, Sabrina Taliani, Federico Da Settimo, Ettore Novellino, Karl Norbert Klotz, Giampiero Spalluto, and Stefano Moro . Revisiting a Receptor-Based Pharmacophore Hypothesis for Human A2A Adenosine Receptor Antagonists. Journal of Chemical Information and Modeling 2013, 53
(7)
, 1620-1637. https://doi.org/10.1021/ci300615u
- Yasushi Yoshikawa, Shinya Oishi, Tatsuhiko Kubo, Noriko Tanahara, Nobutaka Fujii, and Toshio Furuya . Optimized Method of G-Protein-Coupled Receptor Homology Modeling: Its Application to the Discovery of Novel CXCR7 Ligands. Journal of Medicinal Chemistry 2013, 56
(11)
, 4236-4251. https://doi.org/10.1021/jm400307y
- Anne Stößel, Miriam Schlenk, Sonja Hinz, Petra Küppers, Jag Heer, Michael Gütschow, and Christa E. Müller . Dual Targeting of Adenosine A2A Receptors and Monoamine Oxidase B by 4H-3,1-Benzothiazin-4-ones. Journal of Medicinal Chemistry 2013, 56
(11)
, 4580-4596. https://doi.org/10.1021/jm400336x
- Kenneth A. Jacobson . Structure-Based Approaches to Ligands for G-Protein-Coupled Adenosine and P2Y Receptors, from Small Molecules to Nanoconjugates. Journal of Medicinal Chemistry 2013, 56
(10)
, 3749-3767. https://doi.org/10.1021/jm400422s
- Dahlia R. Weiss, SeungKirl Ahn, Maria F. Sassano, Andrew Kleist, Xiao Zhu, Ryan Strachan, Bryan L. Roth, Robert J. Lefkowitz, and Brian K. Shoichet . Conformation Guides Molecular Efficacy in Docking Screens of Activated β-2 Adrenergic G Protein Coupled Receptor. ACS Chemical Biology 2013, 8
(5)
, 1018-1026. https://doi.org/10.1021/cb400103f
- John A. Christopher, Jason Brown, Andrew S. Doré, James C. Errey, Markus Koglin, Fiona H. Marshall, David G. Myszka, Rebecca L. Rich, Christopher G. Tate, Benjamin Tehan, Tony Warne, and Miles Congreve . Biophysical Fragment Screening of the β1-Adrenergic Receptor: Identification of High Affinity Arylpiperazine Leads Using Structure-Based Drug Design. Journal of Medicinal Chemistry 2013, 56
(9)
, 3446-3455. https://doi.org/10.1021/jm400140q
- Ana Negri, Marie-Laure Rives, Michael J. Caspers, Thomas E. Prisinzano, Jonathan A. Javitch, and Marta Filizola . Discovery of a Novel Selective Kappa-Opioid Receptor Agonist Using Crystal Structure-Based Virtual Screening. Journal of Chemical Information and Modeling 2013, 53
(3)
, 521-526. https://doi.org/10.1021/ci400019t
- Thijs Beuming and Woody Sherman . Current Assessment of Docking into GPCR Crystal Structures and Homology Models: Successes, Challenges, and Guidelines. Journal of Chemical Information and Modeling 2012, 52
(12)
, 3263-3277. https://doi.org/10.1021/ci300411b
- Jens Carlsson, Dilip K. Tosh, Khai Phan, Zhan-Guo Gao, and Kenneth A. Jacobson . Structure–Activity Relationships and Molecular Modeling of 1,2,4-Triazoles as Adenosine Receptor Antagonists. ACS Medicinal Chemistry Letters 2012, 3
(9)
, 715-720. https://doi.org/10.1021/ml300097g
- Michael M. Mysinger, Michael Carchia, John. J. Irwin, and Brian K. Shoichet . Directory of Useful Decoys, Enhanced (DUD-E): Better Ligands and Decoys for Better Benchmarking. Journal of Medicinal Chemistry 2012, 55
(14)
, 6582-6594. https://doi.org/10.1021/jm300687e
- Christopher J. Langmead, Stephen P. Andrews, Miles Congreve, James C. Errey, Edward Hurrell, Fiona H. Marshall, Jonathan S. Mason, Christine M. Richardson, Nathan Robertson, Andrei Zhukov, and Malcolm Weir . Identification of Novel Adenosine A2A Receptor Antagonists by Virtual Screening. Journal of Medicinal Chemistry 2012, 55
(5)
, 1904-1909. https://doi.org/10.1021/jm201455y
- Francesca Fanelli and Pier G. De Benedetti . Update 1 of: Computational Modeling Approaches to Structure–Function Analysis of G Protein-Coupled Receptors. Chemical Reviews 2011, 111
(12)
, PR438-PR535. https://doi.org/10.1021/cr100437t
- Chris de Graaf, Albert J. Kooistra, Henry F. Vischer, Vsevolod Katritch, Martien Kuijer, Mitsunori Shiroishi, So Iwata, Tatsuro Shimamura, Raymond C. Stevens, Iwan J. P. de Esch, and Rob Leurs . Crystal Structure-Based Virtual Screening for Fragment-like Ligands of the Human Histamine H1 Receptor. Journal of Medicinal Chemistry 2011, 54
(23)
, 8195-8206. https://doi.org/10.1021/jm2011589
- Miles Congreve, Christopher J. Langmead, Jonathan S. Mason, and Fiona H. Marshall . Progress in Structure Based Drug Design for G Protein-Coupled Receptors. Journal of Medicinal Chemistry 2011, 54
(13)
, 4283-4311. https://doi.org/10.1021/jm200371q
- David Rodríguez, Ángel Piñeiro, and Hugo Gutiérrez-de-Terán . Molecular Dynamics Simulations Reveal Insights into Key Structural Elements of Adenosine Receptors. Biochemistry 2011, 50
(19)
, 4194-4208. https://doi.org/10.1021/bi200100t
- Anette G. Sams, Gitte K. Mikkelsen, Mogens Larsen, Morten Langgård, Mark E. Howells, Tenna J. Schrøder, Lise T. Brennum, Lars Torup, Erling B. Jørgensen, Christoffer Bundgaard, Mads Kreilgård, and Benny Bang-Andersen . Discovery of Phosphoric Acid Mono-{2-[(E/Z)-4-(3,3-dimethyl-butyrylamino)-3,5-difluoro-benzoylimino]-thiazol-3-ylmethyl} Ester (Lu AA47070): A Phosphonooxymethylene Prodrug of a Potent and Selective hA2A Receptor Antagonist. Journal of Medicinal Chemistry 2011, 54
(3)
, 751-764. https://doi.org/10.1021/jm1008659
- Sharangdhar S. Phatak, Edgar A. Gatica, and Claudio N. Cavasotto. Ligand-Steered Modeling and Docking: A Benchmarking Study in Class A G-Protein-Coupled Receptors. Journal of Chemical Information and Modeling 2010, 50
(12)
, 2119-2128. https://doi.org/10.1021/ci100285f
- Miru Tang, Chang Wen, Jie Lin, Hongming Chen, Ting Ran. Discovery of novel A2AR antagonists through deep learning-based virtual screening. Artificial Intelligence in the Life Sciences 2023, 3 , 100058. https://doi.org/10.1016/j.ailsci.2023.100058
- Pierre Matricon, Anh TN. Nguyen, Duc Duy Vo, Jo-Anne Baltos, Mariama Jaiteh, Andreas Luttens, Stefanie Kampen, Arthur Christopoulos, Jan Kihlberg, Lauren Therese May, Jens Carlsson. Structure-based virtual screening discovers potent and selective adenosine A1 receptor antagonists. European Journal of Medicinal Chemistry 2023, 257 , 115419. https://doi.org/10.1016/j.ejmech.2023.115419
- Haruki Yamane, Takashi Ishida. Helix encoder: a compound-protein interaction prediction model specifically designed for class A GPCRs. Frontiers in Bioinformatics 2023, 3 https://doi.org/10.3389/fbinf.2023.1193025
- Chia-Ju Hsieh, Sam Giannakoulias, E. James Petersson, Robert H. Mach. Computational Chemistry for the Identification of Lead Compounds for Radiotracer Development. Pharmaceuticals 2023, 16
(2)
, 317. https://doi.org/10.3390/ph16020317
- Ashraf Ahmed Ali Abdusalam. In-silico identification of novel inhibitors for human Aurora kinase B form the ZINC database using molecular docking-based virtual screening. Research Results in Pharmacology 2022, 8
(4)
, 89-99. https://doi.org/10.3897/rrpharmacology.8.82977
- Kenneth A. Jacobson, Zhan‐Guo Gao, Pierre Matricon, Matthew T. Eddy, Jens Carlsson. Adenosine A
2A
receptor antagonists: from caffeine to selective non‐xanthines. British Journal of Pharmacology 2022, 179
(14)
, 3496-3511. https://doi.org/10.1111/bph.15103
- Miru Tang, Chang Wen, Lin Jie, Hongming Chen, Ting Ran. Discovery of Novel A 2AR Antagonists Through Deep Learning-Based Virtual Screening. SSRN Electronic Journal 2022, 31 https://doi.org/10.2139/ssrn.4188435
- Wen-Ting Chu, Zhiqiang Yan, Xiakun Chu, Xiliang Zheng, Zuojia Liu, Li Xu, Kun Zhang, Jin Wang. Physics of biomolecular recognition and conformational dynamics. Reports on Progress in Physics 2021, 84
(12)
, 126601. https://doi.org/10.1088/1361-6633/ac3800
- Flavio Ballante, Albert J Kooistra, Stefanie Kampen, Chris de Graaf, Jens Carlsson, . Structure-Based Virtual Screening for Ligands of G Protein–Coupled Receptors: What Can Molecular Docking Do for You?. Pharmacological Reviews 2021, 73
(4)
, 1698-1736. https://doi.org/10.1124/pharmrev.120.000246
- Miles Congreve, John A. Christopher, Chris de Graaf. Structure‐Based Drug Design for G Protein‐Coupled Receptors. 2021, 1-59. https://doi.org/10.1002/0471266949.bmc269
- Mukuo Wang, Shujing Hou, Yu Wei, Dongmei Li, Jianping Lin, . Discovery of novel dual adenosine A1/A2A receptor antagonists using deep learning, pharmacophore modeling and molecular docking. PLOS Computational Biology 2021, 17
(3)
, e1008821. https://doi.org/10.1371/journal.pcbi.1008821
- Bassam M. Ayoub, Haidy E. Michel, Shereen Mowaka, Moataz S. Hendy, Mariam M. Tadros. Repurposing of Omarigliptin as a Neuroprotective Agent Based on Docking with A2A Adenosine and AChE Receptors, Brain GLP-1 Response and Its Brain/Plasma Concentration Ratio after 28 Days Multiple Doses in Rats Using LC-MS/MS. Molecules 2021, 26
(4)
, 889. https://doi.org/10.3390/molecules26040889
- Kalpana K. Bhanumathy, Omar Abuhussein, Frederick S. Vizeacoumar, Andrew Freywald, Franco J. Vizeacoumar, Christopher P. Phenix, Eric W. Price, Ran Cao. Computational Prediction of Chemical Tools for Identification and Validation of Synthetic Lethal Interaction Networks. 2021, 333-358. https://doi.org/10.1007/978-1-0716-1740-3_18
- Veronica Salmaso, Kenneth A. Jacobson. Purinergic Signaling: Impact of GPCR Structures on Rational Drug Design. ChemMedChem 2020, 15
(21)
, 1958-1973. https://doi.org/10.1002/cmdc.202000465
- Miles Congreve, Chris de Graaf, Nigel A. Swain, Christopher G. Tate. Impact of GPCR Structures on Drug Discovery. Cell 2020, 181
(1)
, 81-91. https://doi.org/10.1016/j.cell.2020.03.003
- Reed M. Stein, Hye Jin Kang, John D. McCorvy, Grant C. Glatfelter, Anthony J. Jones, Tao Che, Samuel Slocum, Xi-Ping Huang, Olena Savych, Yurii S. Moroz, Benjamin Stauch, Linda C. Johansson, Vadim Cherezov, Terry Kenakin, John J. Irwin, Brian K. Shoichet, Bryan L. Roth, Margarita L. Dubocovich. Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 2020, 579
(7800)
, 609-614. https://doi.org/10.1038/s41586-020-2027-0
- Mariama Jaiteh, Ismael Rodríguez-Espigares, Jana Selent, Jens Carlsson, . Performance of virtual screening against GPCR homology models: Impact of template selection and treatment of binding site plasticity. PLOS Computational Biology 2020, 16
(3)
, e1007680. https://doi.org/10.1371/journal.pcbi.1007680
- Cuihua Zhang, Qunlin Li, Lingwei Meng, Yujie Ren. Design of novel dopamine D
2
and serotonin 5-HT
2A
receptors dual antagonists toward schizophrenia: An integrated study with QSAR, molecular docking, virtual screening and molecular dynamics simulations. Journal of Biomolecular Structure and Dynamics 2020, 38
(3)
, 860-885. https://doi.org/10.1080/07391102.2019.1590244
- Yu Wei, Mukuo Wang, Yang Li, Zhangyong Hong, Dongmei Li, Jianping Lin. Identification of new potent A1 adenosine receptor antagonists using a multistage virtual screening approach. European Journal of Medicinal Chemistry 2020, 187 , 111936. https://doi.org/10.1016/j.ejmech.2019.111936
- Francesca Deflorian, Jonathan S. Mason, Andrea Bortolato, Benjamin G. Tehan. Impact of Recently Determined Crystallographic Structures of GPCRs on Drug Discovery. 2020, 449-477. https://doi.org/10.1002/9781118681121.ch19
- Xiangli Qu, Dejian Wang, Beili Wu. Progress in GPCR structure determination. 2020, 3-22. https://doi.org/10.1016/B978-0-12-816228-6.00001-5
- Jinan Wang, Apurba Bhattarai, Waseem Imtiaz Ahmad, Treyton S. Farnan, Karen Priyadarshini John, Yinglong Miao. Computer-aided GPCR drug discovery. 2020, 283-293. https://doi.org/10.1016/B978-0-12-816228-6.00015-5
- Sumit Jamwal, Ashish Mittal, Puneet Kumar, Dana M. Alhayani, Amal Al-Aboudi. Therapeutic Potential of Agonists and Antagonists of A1, A2a, A2b and A3 Adenosine Receptors. Current Pharmaceutical Design 2019, 25
(26)
, 2892-2905. https://doi.org/10.2174/1381612825666190716112319
- Omar H.A. Al-Attraqchi, Mahesh Attimarad, Katharigatta N. Venugopala, Anroop Nair, Noor H.A. Al-Attraqchi. Adenosine A2A Receptor as a Potential Drug Target - Current Status and Future Perspectives. Current Pharmaceutical Design 2019, 25
(25)
, 2716-2740. https://doi.org/10.2174/1381612825666190716113444
- Pabitra Narayan Samanta, Supratik Kar, Jerzy Leszczynski. Recent Advances of In-Silico Modeling of Potent Antagonists for the Adenosine Receptors. Current Pharmaceutical Design 2019, 25
(7)
, 750-773. https://doi.org/10.2174/1381612825666190304123545
- Nikhil Agrawal, Balakumar Chandrasekaran, Amal Al-Aboudi. Recent Advances in the In-silico Structure-based and Ligand-based Approaches for the Design and Discovery of Agonists and Antagonists of A2A Adenosine Receptor. Current Pharmaceutical Design 2019, 25
(7)
, 774-782. https://doi.org/10.2174/1381612825666190306162006
- H. T. Zhu, L. Y. Qin, T. Liu, Y. Luo. A Convenient Synthesis of N2-Alkylated Guanines. Russian Journal of Organic Chemistry 2019, 55
(6)
, 874-878. https://doi.org/10.1134/S1070428019060198
- Rita C. Acúrcio, Anna Scomparin, Ronit Satchi‐Fainaro, Helena F. Florindo, Rita C. Guedes. Computer‐aided drug design in new druggable targets for the next generation of immune‐oncology therapies. WIREs Computational Molecular Science 2019, 9
(3)
https://doi.org/10.1002/wcms.1397
- Guillaume Ferré, Georges Czaplicki, Pascal Demange, Alain Milon. Structure and dynamics of dynorphin peptide and its receptor. 2019, 17-47. https://doi.org/10.1016/bs.vh.2019.05.006
- Miles Congreve, Giles A. Brown, Alexandra Borodovsky, Michelle L. Lamb. Targeting adenosine A
2A
receptor antagonism for treatment of cancer. Expert Opinion on Drug Discovery 2018, 13
(11)
, 997-1003. https://doi.org/10.1080/17460441.2018.1534825
- Shaherin Basith, Minghua Cui, Stephani J. Y. Macalino, Jongmi Park, Nina A. B. Clavio, Soosung Kang, Sun Choi. Exploring G Protein-Coupled Receptors (GPCRs) Ligand Space via Cheminformatics Approaches: Impact on Rational Drug Design. Frontiers in Pharmacology 2018, 9 https://doi.org/10.3389/fphar.2018.00128
- Magdalena Korczynska, Mary J. Clark, Celine Valant, Jun Xu, Ee Von Moo, Sabine Albold, Dahlia R. Weiss, Hayarpi Torosyan, Weijiao Huang, Andrew C. Kruse, Brent R. Lyda, Lauren T. May, Jo-Anne Baltos, Patrick M. Sexton, Brian K. Kobilka, Arthur Christopoulos, Brian K. Shoichet, Roger K. Sunahara. Structure-based discovery of selective positive allosteric modulators of antagonists for the M
2
muscarinic acetylcholine receptor. Proceedings of the National Academy of Sciences 2018, 115
(10)
https://doi.org/10.1073/pnas.1718037115
- Khushal Kapadiya, Yashwantsinh Jadeja, Ranjan Khunt. Synthesis of Purine‐based Triazoles by Copper (I)‐catalyzed Huisgen Azide–Alkyne Cycloaddition Reaction. Journal of Heterocyclic Chemistry 2018, 55
(1)
, 199-208. https://doi.org/10.1002/jhet.3025
- Agostinho Lemos, Rita Melo, Irina S. Moreira, M. Natália D. S. Cordeiro. Computer-Aided Drug Design Approaches to Study Key Therapeutic Targets in Alzheimer’s Disease. 2018, 61-106. https://doi.org/10.1007/978-1-4939-7404-7_3
- Antonella Ciancetta, Kenneth A. Jacobson. Breakthrough in GPCR Crystallography and Its Impact on Computer-Aided Drug Design. 2018, 45-72. https://doi.org/10.1007/978-1-4939-7465-8_3
- Stefania Baraldi, Pier Giovanni Baraldi, Paola Oliva, Kiran S. Toti, Antonella Ciancetta, Kenneth A. Jacobson. A2A Adenosine Receptor: Structures, Modeling, and Medicinal Chemistry. 2018, 91-136. https://doi.org/10.1007/978-3-319-90808-3_5
- Eric Rouviere, Clément Arnarez, Lewen Yang, Edward Lyman. Identification of Two New Cholesterol Interaction Sites on the A2A Adenosine Receptor. Biophysical Journal 2017, 113
(11)
, 2415-2424. https://doi.org/10.1016/j.bpj.2017.09.027
- Mette Trauelsen, Elisabeth Rexen Ulven, Siv A. Hjorth, Matjaz Brvar, Claudia Monaco, Thomas M. Frimurer, Thue W. Schwartz. Receptor structure-based discovery of non-metabolite agonists for the succinate receptor GPR91. Molecular Metabolism 2017, 6
(12)
, 1585-1596. https://doi.org/10.1016/j.molmet.2017.09.005
- Pierre Matricon, Anirudh Ranganathan, Eugene Warnick, Zhan-Guo Gao, Axel Rudling, Catia Lambertucci, Gabriella Marucci, Aitakin Ezzati, Mariama Jaiteh, Diego Dal Ben, Kenneth A. Jacobson, Jens Carlsson. Fragment optimization for GPCRs by molecular dynamics free energy calculations: Probing druggable subpockets of the A
2A
adenosine receptor binding site. Scientific Reports 2017, 7
(1)
https://doi.org/10.1038/s41598-017-04905-0
- Phillip T. Lowe, Sergio Dall'Angelo, Thea Mulder‐Krieger, Adriaan P. IJzerman, Matteo Zanda, David O'Hagan. A New Class of Fluorinated A
2A
Adenosine Receptor Agonist with Application to Last‐Step Enzymatic [
18
F]Fluorination for PET Imaging. ChemBioChem 2017, 18
(21)
, 2156-2164. https://doi.org/10.1002/cbic.201700382
- Bryan L Roth, John J Irwin, Brian K Shoichet. Discovery of new GPCR ligands to illuminate new biology. Nature Chemical Biology 2017, 13
(11)
, 1143-1151. https://doi.org/10.1038/nchembio.2490
- Sheng Wang, Daniel Wacker, Anat Levit, Tao Che, Robin M. Betz, John D. McCorvy, A. J. Venkatakrishnan, Xi-Ping Huang, Ron O. Dror, Brian K. Shoichet, Bryan L. Roth. D
4
dopamine receptor high-resolution structures enable the discovery of selective agonists. Science 2017, 358
(6361)
, 381-386. https://doi.org/10.1126/science.aan5468
- Valentina Abet, Fabiana Filace, Javier Recio, Julio Alvarez-Builla, Carolina Burgos. Prodrug approach: An overview of recent cases. European Journal of Medicinal Chemistry 2017, 127 , 810-827. https://doi.org/10.1016/j.ejmech.2016.10.061
- Anirudh Ranganathan, David Rodríguez, Jens Carlsson. Structure-Based Discovery of GPCR Ligands from Crystal Structures and Homology Models. 2017, 65-99. https://doi.org/10.1007/7355_2016_25
- M. Congreve, A. Bortolato, G. Brown, R.M. Cooke. Modeling and Design for Membrane Protein Targets. 2017, 145-188. https://doi.org/10.1016/B978-0-12-409547-2.12358-3
- Eelke B. Lenselink, Thijs Beuming, Corine van Veen, Arnault Massink, Woody Sherman, Herman W. T. van Vlijmen, Adriaan P. IJzerman. In search of novel ligands using a structure-based approach: a case study on the adenosine A2A receptor. Journal of Computer-Aided Molecular Design 2016, 30
(10)
, 863-874. https://doi.org/10.1007/s10822-016-9963-7
- Aashish Manglik, Henry Lin, Dipendra K. Aryal, John D. McCorvy, Daniela Dengler, Gregory Corder, Anat Levit, Ralf C. Kling, Viachaslau Bernat, Harald Hübner, Xi-Ping Huang, Maria F. Sassano, Patrick M. Giguère, Stefan Löber, Da Duan, Grégory Scherrer, Brian K. Kobilka, Peter Gmeiner, Bryan L. Roth, Brian K. Shoichet. Structure-based discovery of opioid analgesics with reduced side effects. Nature 2016, 537
(7619)
, 185-190. https://doi.org/10.1038/nature19112
- Celine Lacroix, Inbar Fish, Hayarpi Torosyan, Pranavan Parathaman, John J. Irwin, Brian K. Shoichet, Stephane Angers, . Identification of Novel Smoothened Ligands Using Structure-Based Docking. PLOS ONE 2016, 11
(8)
, e0160365. https://doi.org/10.1371/journal.pone.0160365
- Elham Safarzadeh, Farhad Jadidi-Niaragh, Morteza Motallebnezhad, Mehdi Yousefi. The role of adenosine and adenosine receptors in the immunopathogenesis of multiple sclerosis. Inflammation Research 2016, 65
(7)
, 511-520. https://doi.org/10.1007/s00011-016-0936-z
Abstract
Chart 1
Chart 1. Structures of Known Agonists (1−3) and Antagonists (4−6) of the A2A Adenosine ReceptorFigure 1
Figure 1. Binding mode of the cocrystallized ligand 6 (A) and the predicted binding modes of the seven ligands discovered in the docking screen (B−H). The A2A AR binding site is shown in white ribbons with the side chains of Glu169 and Asn253 in sticks. In (A) the cocrystallized ligand 6 is shown using orange carbon atoms. In (B−H), the crystallographic ligand is shown using blue lines and the docking poses for the ligands are depicted with orange carbon atoms. Black dotted lines indicate hydrogen bonds. The compounds are (B) 7, (C) 8, (D) 9, (E) 10, (F) 11, (G) 12, and (H) 13.
Figure 2
Figure 2. Representative dose−response curves for displacement of binding of the radiolabeled A2A AR agonist 3 by compounds 9, 10, and 11.
Figure 3
Figure 3. Functional assay based on measuring the production of cAMP for 3 (control), a potent A2A AR agonist, with or without 10 μM 9 or 11. The dose−response curve is shifted for both compounds, as expected in the case of competitive antagonistic inhibition. The % activation refers to production of cAMP normalized to the effect of 3 at 100 μM.
References
ARTICLE SECTIONSThis article references 56 other publications.
- 1Overington, J. P.; Al-Lazikani, B.; Hopkins, A. L. How many drug targets are there? Nat. Rev. Drug Discovery 2006, 5, 993– 996Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1Kju7jM&md5=14b5716f4e10b57ae71bf67e52a21929How many drug targets are there?Overington, John P.; Al-Lazikani, Bissan; Hopkins, Andrew L.Nature Reviews Drug Discovery (2006), 5 (12), 993-996CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. For the past decade, the no. of mol. targets for approved drugs has been debated. Here, we reconcile apparently contradictory previous reports into a comprehensive survey, and propose a consensus no. of current drug targets for all classes of approved therapeutic drugs. One striking feature is the relatively const. historical rate of target innovation (the rate at which drugs against new targets are launched); however, the rate of developing drugs against new families is significantly lower. The recent approval of drugs that target protein kinases highlights two addnl. trends: an emerging realization of the importance of polypharmacol., and also the power of a gene-family-led approach in generating novel and important therapies.
- 2Cherezov, V.; Rosenbaum, D. M.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Kuhn, P.; Weis, W. I.; Kobilka, B. K.; Stevens, R. C. High-resolution crystal structure of an engineered human beta(2)-adrenergic G protein-coupled receptor Science 2007, 318, 1258– 1265Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlGmur7I&md5=12c5bacb8464a4b243fe9341192b5b3bHigh-Resolution Crystal Structure of an Engineered Human β2-Adrenergic G Protein-Coupled ReceptorCherezov, Vadim; Rosenbaum, Daniel M.; Hanson, Michael A.; Rasmussen, Soren G. F.; Thian, Foon Sun; Kobilka, Tong Sun; Choi, Hee-Jung; Kuhn, Peter; Weis, William I.; Kobilka, Brian K.; Stevens, Raymond C.; Takeda, S.; Kadowaki, S.; Haga, T.; Takaesu, H.; Mitaku, S.; Fredriksson, R.; Lagerstrom, M. C.; Lundin, L. G.; Schioth, H. B.; Pierce, K. L.; Premont, R. T.; Lefkowitz, R. J.; Lefkowitz, R. J.; Shenoy, S. K.; Rosenbaum, D. M.Science (Washington, DC, United States) (2007), 318 (5854), 1258-1265CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Second extracellular loop, which in the β2-adrenergic receptor contains an unusual pair of disulfide bonds and an extra helix. This loop and the absence Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors constitute the largest family of eukaryotic signal transduction proteins that communicate across the membrane. We report the crystal structure of a human β2-adrenergic receptor-T4 lysozyme fusion protein bound to the partial inverse agonist carazolol at 2.4 angstrom resoln. The structure provides a high-resoln. view of a human G protein-coupled receptor bound to a diffusible ligand. Ligand-binding site accessibility is enabled by the second extracellular loop, which is held out of the binding cavity by a pair of closely spaced disulfide bridges and a short helical segment within the loop. Cholesterol, a necessary component for crystn., mediates an intriguing parallel assocn. of receptor mols. in the crystal lattice. Although the location of carazolol in the β2-adrenergic receptor is very similar to that of retinal in rhodopsin, structural differences in the ligand-binding site and other regions highlight the challenges in using rhodopsin as a template model for this large receptor family.
- 3Rosenbaum, D. M.; Cherezov, V.; Hanson, M. A.; Rasmussen, S. G. F.; Thian, F. S.; Kobilka, T. S.; Choi, H. J.; Yao, X. J.; Weis, W. I.; Stevens, R. C.; Kobilka, B. K. GPCR engineering yields high-resolution structural insights into beta(2)-adrenergic receptor function Science 2007, 318, 1266– 1273Google ScholarThere is no corresponding record for this reference.
- 4Warne, T.; Serrano-Vega, M. J.; Baker, J. G.; Moukhametzianov, R.; Edwards, P. C.; Henderson, R.; Leslie, A. G. W.; Tate, C. G.; Schertler, G. F. X. Structure of a beta(1)-adrenergic G-protein-coupled receptor Nature 2008, 454, 486– 491Google ScholarThere is no corresponding record for this reference.
- 5Jaakola, V. P.; Griffith, M. T.; Hanson, M. A.; Cherezov, V.; Chien, E. Y. T.; Lane, J. R.; IJzerman, A. P.; Stevens, R. C. The 2.6 angstrom crystal structure of a human A(2A) adenosine receptor bound to an antagonist Science 2008, 322, 1211– 1217Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlyqtbfN&md5=5bdb862b41f345c244f3c162e058206bThe 2.6 Angstrom Crystal Structure of a Human A2A Adenosine Receptor Bound to an AntagonistJaakola, Veli-Pekka; Griffith, Mark T.; Hanson, Michael A.; Cherezov, Vadim; Chien, Ellen Y. T.; Lane, J. Robert; IJzerman, Adriaan P.; Stevens, Raymond C.Science (Washington, DC, United States) (2008), 322 (5905), 1211-1217CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The adenosine class of heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) mediates the important role of extracellular adenosine in many physiol. processes and is antagonized by caffeine. The authors have detd. the crystal structure of the human A2A adenosine receptor, in complex with a high-affinity subtype-selective antagonist, ZM241385, to 2.6 angstrom resoln. Four disulfide bridges in the extracellular domain, combined with a subtle repacking of the transmembrane helixes relative to the adrenergic and rhodopsin receptor structures, define a pocket distinct from that of other structurally detd. GPCRs. The arrangement allows for the binding of the antagonist in an extended conformation, perpendicular to the membrane plane. The binding site highlights an integral role for the extracellular loops, together with the helical core, in ligand recognition by this class of GPCRs and suggests a role for ZM241385 in restricting the movement of a tryptophan residue important in the activation mechanism of the class A receptors.
- 6Congreve, M.; Marshall, F. The impact of GPCR structures on pharmacology and structure-based drug design Br. J. Pharmacol. 2010, 159, 986– 996Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1ektro%253D&md5=e9ab946b907eb44edf516eb3231e6931The impact of GPCR structures on pharmacology and structure-based drug designCongreve, Miles; Marshall, FionaBritish Journal of Pharmacology (2010), 159 (5), 986-996CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)A review. After many years of effort, recent tech. breakthroughs have enabled the X-ray crystal structures of three G-protein-coupled receptors (GPCRs) (β1 and β2 adrenergic and adenosine A2a) to be solved in addn. to rhodopsin. GPCRs, like other membrane proteins, have lagged behind sol. drug targets such as kinases and proteases in the no. of structures available and the level of understanding of these targets and their interaction with drugs. The availability of increasing nos. of structures of GPCRs is set to greatly increase our understanding of some of the key issues in GPCR biol. In particular, what constitutes the different receptor conformations that are involved in signaling and the mol. changes which occur upon receptor activation. How future GPCR structures might alter our views on areas such as agonist-directed signaling and allosteric regulation as well as dimerization is discussed. Knowledge of crystal structures in complex with small mols. will enable techniques in drug discovery and design, which have previously only been applied to sol. targets, to now be used for GPCR targets. These methods include structure-based drug design, virtual screening and fragment screening. This review considers how these methods have been used to address problems in drug discovery for kinase and protease targets and therefore how such methods are likely to impact GPCR drug discovery in the future.
- 7Moro, S.; Gao, Z. G.; Jacobson, K. A.; Spalluto, G. Progress in the pursuit of therapeutic adenosine receptor antagonists Med. Res. Rev. 2006, 26, 131– 159Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XislOksLw%253D&md5=48f7a603f3c4044b2c48aba13a031e07Progress in the pursuit of therapeutic Adenosine receptor antagonistsMoro, Stefano; Gao, Zhan-Guo; Jacobson, Kenneth A.; Spalluto, GiampieroMedicinal Research Reviews (2006), 26 (2), 131-159CODEN: MRREDD; ISSN:0198-6325. (John Wiley & Sons, Inc.)A review. Ever since the discovery of the hypotensive and bradycardiac effects of adenosine, adenosine receptors continue to represent promising drug targets. First, this is due to the fact that the receptors are expressed in a large variety of tissues. In particular, the actions of adenosine (or methylxanthine antagonists) in the central nervous system, in the circulation, on immune cells, and on other tissues can be beneficial in certain disorders. Second, there exists a large no. of ligands, which have been generated by introducing several modifications in the structure of the lead compds. (adenosine and methylxanthine), some of them highly specific. Four adenosine receptor subtypes (A1, A2A, A2B, and A3) have been cloned and pharmacol. characterized, all of which are G protein-coupled receptors. Adenosine receptors can be distinguished according to their preferred mechanism of signal transduction: A1 and A3 receptors interact with pertussis toxin-sensitive G proteins of the Gi and Go family; the canonical signaling mechanism of the A2A and of the A2B receptors is stimulation of adenylyl cyclase via Gs proteins. In addn. to the coupling to adenylyl cyclase, all four subtypes may pos. couple to phospholipase C via different G protein subunits. The development of new ligands, in particular, potent and selective antagonists, for all subtypes of adenosine receptors has so far been directed by traditional medicinal chem. The availability of genetic information promises to facilitate understanding of the drug-receptor interaction leading to the rational design of a potentially therapeutically important class of drugs. Moreover, mol. modeling may further rationalize obsd. interactions between the receptors and their ligands. In this review, we will summarize the most relevant progress in developing new therapeutic adenosine receptor antagonists.
- 8Jacobson, K. A.; Gao, Z. G. Adenosine receptors as therapeutic targets Nat. Rev. Drug Discovery 2006, 5, 247– 264Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhvFOlsr4%253D&md5=85ad553154bd61f24a6bb594f960f9bcAdenosine receptors as therapeutic targetsJacobson, Kenneth A.; Gao, Zhan-GuoNature Reviews Drug Discovery (2006), 5 (3), 247-264CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Adenosine receptors are major targets of caffeine, the most commonly consumed drug in the world. There is growing evidence that they could also be promising therapeutic targets in a wide range of conditions, including cerebral and cardiac ischemic diseases, sleep disorders, immune and inflammatory disorders and cancer. After more than three decades of medicinal chem. research, a considerable no. of selective agonists and antagonists of adenosine receptors have been discovered, and some have been clin. evaluated, although none has yet received regulatory approval. However, recent advances in the understanding of the roles of the various adenosine receptor subtypes, and in the development of selective and potent ligands, as discussed in this review, have brought the goal of therapeutic application of adenosine receptor modulators considerably closer.
- 9Sebastiao, A. M.; Ribeiro, J. A. Adenosine receptors and the central nervous system Handb. Exp. Pharmacol. 2009, 471– 534Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sktr3N&md5=f1c88cee280ebf7783da9efe18eb099eAdenosine receptors and the central nervous systemSebastiao, Ana M.; Ribeiro, Joaquim A.Handbook of Experimental Pharmacology (2009), 193 (Adenosine Receptors in Health and Disease), 471-534CODEN: HEPHD2; ISSN:0171-2004. (Springer GmbH)A review. The adenosine receptors (ARs) in the nervous system act as a kind of "go-between" to regulate the release of neurotransmitters (this includes all known neurotransmitters) and the action of neuromodulators (e.g., neuropeptides, neurotrophic factors). Receptor-receptor interactions and AR-transporter interplay occur as part of the adenosine's attempt to control synaptic transmission. A2AARs are more abundant in the striatum and A1ARs in the hippocampus, but both receptors interfere with the efficiency and plasticity-regulated synaptic transmission in most brain areas. The omnipresence of adenosine and A2A and A1 ARs in all nervous system cells (neurons and glia), together with the intensive release of adenosine following insults, makes adenosine a kind of "maestro" of the tripartite synapse in the homeostatic coordination of the brain function. Under physiol. conditions, both A2A and A1 ARs play an important role in sleep and arousal, cognition, memory and learning, whereas under pathol. conditions (e.g., Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, stroke, epilepsy, drug addiction, pain, schizophrenia, depression), ARs operate a time/circumstance window where in some circumstances A1AR agonists may predominate as early neuroprotectors, and in other circumstances A2AAR antagonists may alter the outcomes of some of the pathol. deficiencies. In some circumstances, and depending on the therapeutic window, the use of A2AAR agonists may be initially beneficial; however, at later time points, the use of A2AAR antagonists proved beneficial in several pathologies. Since selective ligands for A1 and A2A ARs are now entering clin. trials, the time has come to det. the role of these receptors in neurol. and psychiatric diseases and identify therapies that will alter the outcomes of these diseases, therefore providing a hopeful future for the patients who suffer from these diseases.
- 10Blackburn, M. R.; Vance, C. O.; Morschl, E.; Wilson, C. N. Adenosine receptors and inflammation Handb. Exp. Pharmacol. 2009, 215– 269Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sktr%252FK&md5=266c99602d3e0ee0dcfc81a4dfefef47Adenosine receptors and inflammationBlackburn, Michael R.; Vance, Constance O.; Morschl, Eva; Wilson, Constance N.Handbook of Experimental Pharmacology (2009), 193 (Adenosine Receptors in Health and Disease), 215-269CODEN: HEPHD2; ISSN:0171-2004. (Springer GmbH)A review. Extracellular adenosine is produced in a coordinated manner from cells following cellular challenge or tissue injury. Once produced, it serves as an autocrine- and paracrine-signaling mol. through its interactions with seven-membrane-spanning G-protein-coupled adenosine receptors. These signaling pathways have widespread physiol. and pathophysiol. functions. Immune cells express adenosine receptors and respond to adenosine or adenosine agonists in diverse manners. Extensive in vitro and in vivo studies have identified potent anti-inflammatory functions for all of the adenosine receptors on many different inflammatory cells and in various inflammatory disease processes. In addn., specific proinflammatory functions have also been ascribed to adenosine receptor activation. The potent effects of adenosine signaling on the regulation of inflammation suggest that targeting specific adenosine receptor activation or inactivation using selective agonists and antagonists could have important therapeutic implications in numerous diseases. This review is designed to summarize the current status of adenosine receptor signaling in various inflammatory cells and in models of inflammation, with an emphasis on the advancement of adenosine-based therapeutics to treat inflammatory disorders.
- 11Cristalli, G.; Muller, C. E.; Volpini, R. Recent developments in adenosine A2A receptor ligands Handb. Exp. Pharmacol. 2009, 59– 98Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sktr7E&md5=95c3e8fde30c9094d81cea882987ae64Recent developments in adenosine A2A receptor ligandsCristalli, Gloria; Muller, Christa E.; Volpini, RosariaHandbook of Experimental Pharmacology (2009), 193 (Adenosine Receptors in Health and Disease), 59-98CODEN: HEPHD2; ISSN:0171-2004. (Springer GmbH)A review. The development of potent and selective agonists and antagonists of adenosine receptors (ARs) has been a target of medicinal chem. research for several decades, and recently the US Food and Drug Administration has approved Lexiscan, an adenosine deriv. substituted at the 2 position, for use as a pharmacol. stress agent in radionuclide myocardial perfusion imaging. Currently, some other adenosine A2A receptor (A2AAR) agonists and antagonists are undergoing preclin. testing and clin. trials. While agonists are potent antiinflammatory agents also showing hypotensive effects, antagonists are being developed for the treatment of Parkinson's disease. However, since there are still major problems in this field, including side effects, low brain penetration (for the targeting of CNS diseases), short half-life, or lack of in vivo effects, the design and development of new AR ligands is a hot research topic. This review presents an update on the medicinal chem. of A2AAR agonists and antagonists, and stresses the strong need for more selective ligands at the human A2AAR subtype, in particular in the case of agonists.
- 12Poucher, S. M.; Keddie, J. R.; Singh, P.; Stoggall, S. M.; Caulkett, P. W. R.; Jones, G.; Collis, M. G. The in-vitro pharmacology of Zm-241385, a potent, nonxanthine, a(2a) selective adenosine receptor antagonist Br. J. Pharmacol. 1995, 115, 1096– 1102Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXnt1Gksrk%253D&md5=d0f1f44a790e48e5c2c04fe0d71e3638The in vitro pharmacology of ZM 241385, a potent, non-xanthine, A2a selective adenosine receptor antagonistPoucher, S. M.; Keddie, J. R.; Singh, P.; Stoggall, S. M.; Caulkett, P. W. R.; Jones, G.; Collis, M. G.British Journal of Pharmacology (1995), 115 (6), 1096-102CODEN: BJPCBM; ISSN:0007-1188. (Macmillan Scientific & Medical Division)This paper describes the in vitro pharmacol. of ZM 241385 (4-(2-[7-amino-2-(2-furyl) [1,2,4]-triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol), a novel non-xanthine adenosine receptor antagonist with selectivity for the A2 receptor subtype. ZM 241385 had high affinity for A2a receptors. In rat pheochromocytoma cell membranes, ZM 241385 displaced binding of tritiated 5'-N-ethylcarboxamidoadenosine (NECA) with a pIC50 of 9.52, (95% confidence limits, c.l., 9.02-10.02). In guinea-pig isolated Langendorff hearts, ZM 241385 antagonized vasodilation of the coronary bed produced by 2-chloroadenosine (2-CADO) and 2-[p-(2-carboxyethyl) phenylamino]-5'-N-ethylcarboxamidoadenosine (CGS21680) with pA2 values of 8.57 (c.l., 8.45-8.68) and 9.02 (c.l., 8.79-9.24) resp. ZM 241385 had low potency at A2b receptors and antagonized the relaxant effects of adenosine in the guinea-pig aorta with a pA2 of 7.06 (c.l., 6.92-7.19). ZM 241385 had a low affinity at A1 receptors. In rat cerebral cortex membranes it displayed tritiated R-phenylisopropyladenosine (R-PIA) with a pIC50 of 5.69 (c.l., 5.57-5.81). ZM 241385 antagonized the bradycardic action of 2-CADO in guinea-pig atria with a pA2 of 5.95 (c.l., 5.72-6.18). ZM 241385 had low affinity for A3 receptors. At cloned rat A3 receptors expressed in chinese hamster ovary cells, it displayed iodinated aminobenzyl-5'-N-methylcarboxamido adenosine (AB-MECA) with a pIC50 of 3.82 (c.l., 3.67-4.06). ZM 241385 had no significant addnl. pharmacol. effects on the isolated tissues used in these studies at concns. three orders of magnitude greater than those which block A2a receptors. At 10 μM it displayed only minor inhibition of the bradycardic effects in guinea-pig atria to some concns. of carbachol. At 10 μM, ZM 241385 had a small inhibitory effect on relaxant effects of isoprenaline in guinea-pig aorta but no effect on sodium nitrite-induced relaxation. ZM 241385 (100 μM) was without effect on phenylephrine-induced tone in guinea-pig aorta. ZM 241385 (10 μM) had no inhibitory effect on rat hepatocyte phosphodiesterase types I, II, III and IV but caused a small inhibition of the calcium calmodulin-activated type I enzyme. ZM 241385 is the most selective adenosine A2a receptor antagonist yet described and is therefore a useful tool for characterization of responses mediated by A2 adenosine receptors.
- 13Degen, J.; Rarey, M. FlexNovo: structure-based searching in large fragment spaces ChemMedChem 2006, 1, 854– 868Google ScholarThere is no corresponding record for this reference.
- 14Jones, G.; Willett, P.; Glen, R. C.; Leach, A. R.; Taylor, R. Development and validation of a genetic algorithm for flexible docking J. Mol. Biol. 1997, 267, 727– 748Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXis1KntLo%253D&md5=476a2b1d8f80f3ba418052fe29d735caDevelopment and validation of a genetic algorithm for flexible dockingJones, Gareth; Willett, Peter; Glen, Robert C.; Leach, Andrew R.; Taylor, RobinJournal of Molecular Biology (1997), 267 (3), 727-748CODEN: JMOBAK; ISSN:0022-2836. (Academic)Prediction of small mol. binding modes to macromols. of known three-dimensional structure is a problem of paramount importance in rational drug design (the "docking" problem). We report the development and validation of the program GOLD (Genetic Optimization for Ligand Docking). GOLD is an automated ligand docking program that uses a genetic algorithm to explore the full range of ligand conformational flexibility with partial flexibility of the protein and satisfies the fundamental requirement that the ligand must displace loosely bound water on binding. Numerous enhancements and modifications have been applied to the original technique resulting in a substantial increase in the reliability and the applicability of the algorithm. The advanced algorithm has been tested on a dataset of 100 complexes extd. from the Brookhaven Protein Data Bank. When used to dock the ligand back into the binding site, GOLD achieved a 71% success rate in identifying the exptl. binding mode.
- 15Kairys, V.; Fernandes, M. X.; Gilson, M. K. Screening drug-like compounds by docking to homology models: a systematic study J. Chem. Inf. Model. 2006, 46, 365– 379Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht12rtbzL&md5=81e8af8b023309a765cff3e9918a4d4cScreening Drug-Like Compounds by Docking to Homology Models: A Systematic StudyKairys, Visvaldas; Fernandes, Miguel X.; Gilson, Michael K.Journal of Chemical Information and Modeling (2006), 46 (1), 365-379CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In the absence of an exptl. solved structure, a homol. model of a protein target can be used instead for virtual screening of drug candidates by docking and scoring. This approach poses a no. of questions regarding the choice of the template to use in constructing the model, the accuracy of the screening results, and the importance of allowing for protein flexibility. The present study addresses such questions with compd. screening calcns. for multiple homol. models of five drug targets. A central result is that docking to homol. models frequently yields enrichments of known ligands as good as that obtained by docking to a crystal structure of the actual target protein. Interestingly, however, std. measures of the similarity of the template used to build the homol. model to the targeted protein show little correlation with the effectiveness of the screening calcns., and docking to the template itself often is as successful as docking to the corresponding homol. model. Treating key side chains as mobile produces a modest improvement in the results. The reasons for these sometimes unexpected results, and their implications for future methodol. development, are discussed.
- 16Lorber, D. M.; Shoichet, B. K. Flexible ligand docking using conformational ensembles Protein Sci. 1998, 7, 938– 950Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXisFSnurg%253D&md5=64d79dfe6a048c76aec43f8bd9f6bc62Flexible ligand docking using conformational ensemblesLorber, David M.; Shoichet, Brian K.Protein Science (1998), 7 (4), 938-950CODEN: PRCIEI; ISSN:0961-8368. (Cambridge University Press)Mol. docking algorithms suggest possible structures for mol. complexes. They are used to model biol. function and to discover potential ligands. A present challenge for docking algorithms is the treatment of mol. flexibility. Here, the rigid body program, DOCK, is modified to allow it to rapidly fit multiple conformations of ligands. Conformations of a given mol. are pre-calcd. in the same frame of ref., so that each conformer shares a common rigid fragment with all other conformations. The ligand conformers are then docked together, as an ensemble, into a receptor binding site. This takes advantage of the redundancy present in differing conformers of the same mol. The algorithm was tested using three org. ligand protein systems and two protein-protein systems. Both the bound and unbound conformations of the receptors were used. The ligand ensemble method found conformations that resembled those detd. in X-ray crystal structures (RMS values typically less than 1.5 Å). To test the method's usefulness for inhibitor discovery, multi-compd. and multi-conformer databases were screened for compds. known to bind to dihydrofolate reductase and compds. known to bind to thymidylate synthase. In both cases, known inhibitors and substrates were identified in conformations resembling those obsd. exptl. The ligand ensemble method was 100-fold faster than docking a single conformation at a time and was able to screen a database of over 34 million conformations from 117,000 mols. in one to four CPU days on a workstation.
- 17Lorber, D. M.; Shoichet, B. K. Hierarchical docking of databases of multiple ligand conformations Curr. Top. Med. Chem. 2005, 5, 739– 749Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpsFGht74%253D&md5=25c833b5608e5faedc1af3ad38fdfa06Hierarchical docking of databases of multiple ligand conformationsLorber, David M.; Shoichet, Brian K.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2005), 5 (8), 739-749CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)Ligand flexibility is an important problem in mol. docking and virtual screening. To address this challenge, we investigate a hierarchical pre-organization of multiple conformations of small mols. Such organization of pre-calcd. conformations removes the exploration of ligand conformational space from the docking calcn. and allows for concise representation of what can be thousands of conformations. The hierarchy also recognizes and prunes incompatible conformations early in the calcn., eliminating redundant calcns. of fit. We investigate the method by docking the MDL Drug Data Report (MDDR), an annotated database of 100,000 mols., into apo and holo forms of 7 unrelated targets. This annotated database allows us to track the ranking of tens to hundreds of annotated ligands in each of the docking systems. The binding sites and database are prepd. in an automated fashion in an attempt to remove some human bias from the calcns. Many thousands of explicit and implicit ligand conformations may be docked in calcns. not much longer than required for single conformer docking. As long as internal energies are not considered, recombination with the hierarchy is additive as the no. of degrees of freedom is increased. Mols. with even millions of conformations can be docked in a few minutes on a single desktop computer.
- 18Zavodszky, M. I.; Kuhn, L. A. Side-chain flexibility in protein−ligand binding: the minimal rotation hypothesis Protein Sci. 2005, 14, 1104– 1114Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXislOmsrc%253D&md5=1cfcffacb29da4ab665de67b2189dc4eSide-chain flexibility in protein-ligand binding: The minimal rotation hypothesisZavodszky, Maria I.; Kuhn, Leslie A.Protein Science (2005), 14 (4), 1104-1114CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)The goal of this work is to learn from nature about the magnitudes of side-chain motions that occur when proteins bind small org. mols. and model these motions to improve the prediction of protein-ligand complexes. Following anal. of protein side-chain motions upon ligand binding in 63 complexes, we tested the ability of the docking tool SLIDE to model these motions without being restricted to rotameric transitions or deciding which side chains should be considered as flexible. The model tested is that side-chain conformational changes involving more atoms or larger rotations are likely to be more costly and less prevalent than small motions due to energy barriers between rotamers and the potential of large motions to cause new steric clashes. Accordingly, SLIDE adjusts the protein and ligand side groups as little as necessary to achieve steric complementarity. We tested the hypothesis that small motions are sufficient to achieve good dockings using 63 ligands and the apo structures of 20 different proteins and compared SLIDE side-chain rotations to those exptl. obsd. None of these proteins undergoes major main-chain conformational change upon ligand binding, ensuring that side-chain flexibility modeling is not required to compensate for main-chain motions. Although more frugal in the no. of side-chain rotations performed, this model substantially mimics the exptl. obsd. motions. Most side chains do not shift to a new rotamer, and small motions are both necessary and sufficient to predict the correct binding orientation and most protein-ligand interactions for the 20 proteins analyzed.
- 19Kolb, P.; Rosenbaum, D. M.; Irwin, J. J.; Fung, J. J.; Kobilka, B. K.; Shoichet, B. K. Structure-based discovery of beta(2)-adrenergic receptor ligands Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 6843– 6848Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlsV2qsro%253D&md5=4d1a4cb2aa3925aa4c99c6b0496417a7Structure-based discovery of β2-adrenergic receptor ligandsKolb, Peter; Rosenbaum, Daniel M.; Irwin, John J.; Fung, Juan Jose; Kobilka, Brian K.; Shoichet, Brian K.Proceedings of the National Academy of Sciences of the United States of America (2009), 106 (16), 6843-6848CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Aminergic G protein-coupled receptors (GPCRs) have been a major focus of pharmaceutical research for many years. Due partly to the lack of reliable receptor structures, drug discovery efforts have been largely ligand-based. The recently detd. X-ray structure of the β2-adrenergic receptor offers an opportunity to investigate the advantages and limitations inherent in a structure-based approach to ligand discovery against this and related GPCR targets. Approx. 1 million com. available, "lead-like" mols. were docked against the β2-adrenergic receptor structure. On testing of 25 high-ranking mols., 6 were active with binding affinities <4 μM, with the best mol. binding with a Ki of 9 nM (95% confidence interval 7-10 nM). Five of these mols. were inverse agonists. The high hit rate, the high affinity of the most potent mol., the discovery of unprecedented chemotypes among the new inhibitors, and the apparent bias toward inverse agonists among the docking hits, have implications for structure-based approaches against GPCRs that recognize small org. mols.
- 20Sabio, M.; Jones, K.; Topiol, S. Use of the X-ray structure of the beta(2)-adrenergic receptor for drug discovery. Part 2: Identification of active compounds Bioorg. Med. Chem. Lett. 2008, 18, 5391– 5395Google ScholarThere is no corresponding record for this reference.
- 21de Graaf, C.; Rognan, D. Selective structure-based virtual screening for full and partial agonists of the beta 2 adrenergic receptor J. Med. Chem. 2008, 51, 4978– 4985Google ScholarThere is no corresponding record for this reference.
- 22Katritch, V.; Reynolds, K. A.; Cherezov, V.; Hanson, M. A.; Roth, C. B.; Yeager, M.; Abagyan, R. Analysis of full and partial agonists binding to beta(2)-adrenergic receptor suggests a role of transmembrane helix V in agonist-specific conformational changes J. Mol. Recognit. 2009, 22, 307– 318Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXnslGis7k%253D&md5=01e2c841b200697382e551b3cf104451Analysis of full and partial agonists binding to β2-adrenergic receptor suggests a role of transmembrane helix V in agonist-specific conformational changesKatritch, Vsevolod; Reynolds, Kimberly A.; Cherezov, Vadim; Hanson, Michael A.; Roth, Christopher B.; Yeager, Mark; Abagyan, RubenJournal of Molecular Recognition (2009), 22 (4), 307-318CODEN: JMORE4; ISSN:0952-3499. (John Wiley & Sons Ltd.)The 2.4 Å crystal structure of the β2-adrenergic receptor (β2AR) in complex with the high-affinity inverse agonist (-)-carazolol provides a detailed structural framework for the anal. of ligand recognition by adrenergic receptors. Insights into agonist binding and the corresponding conformational changes triggering G-protein coupled receptor (GPCR) activation mechanism are of special interest. While the carazolol pocket captured in the β2AR crystal structure accommodates (-)-isoproterenol and other agonists without steric clashes, a finite movement of the flexible extracellular part of TM-V helix (TM-Ve) obtained by receptor optimization in the presence of docked ligand can further improve the calcd. binding affinities for agonist compds. Tilting of TM-Ve towards the receptor axis provides a more complete description of polar receptor-ligand interactions for full and partial agonists, by enabling optimal engagement of agonists with two exptl. identified anchor sites, formed by Asp 113/Asn 312 and Ser 203/Ser 204/Ser 207 side chains. Further, receptor models incorporating a flexible TM-V backbone allow reliable prediction of binding affinities for a set of diverse ligands, suggesting potential utility of this approach to design of effective and subtype-specific agonists for adrenergic receptors. Systematic differences in capacity of partial, full and inverse agonists to induce TM-V helix tilt in the β2AR model suggest potential role of TM-V as a conformational "rheostat" involved in the whole spectrum of β2AR responses to small mol. signals.
- 23Reynolds, K. A.; Katritch, V.; Abagyan, R. Identifying conformational changes of the beta(2) adrenoceptor that enable accurate prediction of ligand/receptor interactions and screening for GPCR modulators J. Comput.-Aided Mol. Des. 2009, 23, 273– 288Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXkt1yitLw%253D&md5=48e809569368d89fb9ba2c36bddc3f3bIdentifying conformational changes of the β2 adrenoceptor that enable accurate prediction of ligand/receptor interactions and screening for GPCR modulatorsReynolds, Kimberly A.; Katritch, Vsevolod; Abagyan, RubenJournal of Computer-Aided Molecular Design (2009), 23 (5), 273-288CODEN: JCADEQ; ISSN:0920-654X. (Springer)The new β2 Adrenoceptor (β2AR) crystal structures provide a high-resoln. snapshot of receptor interactions with two particular partial inverse agonists, (-)-carazolol and timolol. However, both exptl. and computational studies of GPCR structure are significantly complicated by the existence of multiple conformational states coupled to ligand type and receptor activity. Agonists and antagonists induce or stabilize distinct changes in receptor structure that mediate a range of pharmacol. activities. In this work, the authors (1) established that the existing β2AR crystallog. conformers can be extended to describe ligand/receptor interactions for addnl. antagonist types, (2) generated agonist-bound receptor conformations, and (3) validated these models for agonist and antagonist virtual ligand screening (VLS). Using a ligand directed refinement protocol, the authors derived a single agonist-bound receptor conformation that selectively retrieved a diverse set of full and partial β2AR agonists in VLS trials. Addnl., the impact of extracellular loop two conformation on VLS was assessed by docking studies with rhodopsin-based β2AR homol. models, and loop-deleted receptor models. A general strategy for constructing and selecting agonist-bound receptor pocket conformations is presented, which may prove broadly useful in creating agonist and antagonist bound models for other GPCRs.
- 24Kuntz, I. D.; Blaney, J. M.; Oatley, S. J.; Langridge, R.; Ferrin, T. E. A geometric approach to macromolecule−ligand interactions J. Mol. Biol. 1982, 161, 269– 288Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL38XmtFajsbw%253D&md5=8a4234b24356ea5340f33d906cd71d3eA geometric approach to macromolecule-ligand interactionsKuntz, Irwin D.; Blaney, Jeffrey M.; Oatley, Stuart J.; Langridge, Robert; Ferrin, Thomas E.Journal of Molecular Biology (1982), 161 (2), 269-88CODEN: JMOBAK; ISSN:0022-2836.A method is described to explore geometrically feasible alignments of ligands and receptors of known structure. Algorithms are presented that examine many binding geometries and evaluate them in terms of steric overlap. The procedure uses specific mol. conformations. A method is included for finding putative binding sites on a macromol. surface. Results are reported for heme-myoglobin interaction and the binding of thyroid hormone analogs to prealbumin. In each case, the program finds structures within 1 Å of the x-ray results and also finds distinctly different geometries that provide good steric fits. The approach seems well-suited for generating conformations for energy refinement programs and interactive computer graphics routines.
- 25Shoichet, B. K.; Kuntz, I. D. Matching chemistry and shape in molecular docking Protein Eng. 1993, 6, 723– 732Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXnt1yhtQ%253D%253D&md5=d2ed691493e3dd8701fc19ddd1e7af66Matching chemistry and shape in molecular dockingShoichet, Brian K.; Kuntz, Irwin D.Protein Engineering (1993), 6 (7), 723-32CODEN: PRENE9; ISSN:0269-2139.The authors have added a chem. filter to the ligand placement algorithm of the mol. docking program DOCK. DOCK places ligands in receptors using local shape features. Here the authors label these shape features by chem. type and insist on complementary matches. The authors find fewer phys. unrealistic complexes without reducing the no. of complexes resembling the known ligand-receptor configurations. Approx. 10-fold fewer complexes are calcd. and the new algorithm is correspondingly 10-fold faster than the previous shape-only matching. The authors tested the new algorithm's ability to reproduce three known ligand-receptor complexes: methotrexate in dihydrofolate reductase, deoxyuridine monophosphate in thymidylate synthase and pancreatic trypsin inhibitor in trypsin. The program found configurations within 1 Å of the crystallog. mode, with fewer nonnative solns. compared with shape-only matching. The authors also tested the program's ability to retrieve known inhibitors of thymidylate synthase and dihydrofolate reductase by screening mol. databases against the enzyme structures. Both algorithms retrieved many known inhibitors preferentially to other compds. in the database. The chem. matching algorithm generally ranks known inhibitors better than does matching based on shape alone.
- 26Nicholls, A.; Honig, B. A rapid finite-difference algorithm, utilizing successive over-relaxation to solve the Poisson−Boltzmann equation J. Comput. Chem. 1991, 12, 435– 445Google ScholarThere is no corresponding record for this reference.
- 27Weiner, S. J.; Kollman, P. A.; Case, D. A.; Singh, U. C.; Ghio, C.; Alagona, G.; Profeta, S.; Weiner, P. A new force-field for molecular mechanical simulation of nucleic-acids and proteins J. Am. Chem. Soc. 1984, 106, 765– 784Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXmsVCgug%253D%253D&md5=c4aa27feec7e2c4d34001be10a6cb8e9A new force field for molecular mechanical simulation of nucleic acids and proteinsWeiner, Scott J.; Kollman, Peter A.; Case, David A.; Singh, U. Chandra; Ghio, Caterina; Alagona, Guliano; Profeta, Salvatore, Jr.; Weiner, PaulJournal of the American Chemical Society (1984), 106 (3), 765-84CODEN: JACSAT; ISSN:0002-7863.A force field was developed for simulation of nucleic acids and proteins. The approach began by obtaining equil. bond lengths and angles from microwave, neutron diffraction, and prior mol. mech. calcns., torsional consts. from microwave, NMR, and mol. mech. studies, nonbonded parameters from crystal packing calcns., and at. charges from the fit of a partial charge model to electrostatic potentials calcd. by ab initio quantum mech. theory. The parameters then were refined with mol. mech. studies on the structures and energies of model compds. For nucleic acids, Me Et ether, THF, deoxyadenosine, di-Me phosphate, 9-methylguanine-1-methylcytosine H-bonded complex, 9-methyladenine-1-methylthymine H-bonded complex, and 1,3-dimethyluracil base-stacked dimer were studied. Bond, angle, torsional, nonbonded, and H-bond parameters were varied to optimize the agreement between calcd. and exptl. values for sugar pucker energies and structures, vibrational frequencies of di-Me phosphate and THF, and energies for base pairing and base stacking. For proteins, Φ,ψ maps of glycyl and alanyl dipeptides, H-bonding interactions involving the various protein polar groups, and energy refinement calcns. on insulin were considered. Unlike the models for H bonding involving N and O electron donors, an adequate description of S-H bonding required explicit inclusion of lone pairs.
- 28Babaoglu, K.; Simeonov, A.; Lrwin, J. J.; Nelson, M. E.; Feng, B.; Thomas, C. J.; Cancian, L.; Costi, M. P.; Maltby, D. A.; Jadhav, A.; Inglese, J.; Austin, C. P.; Shoichet, B. K. Comprehensive mechanistic analysis of hits from high-throughput and docking screens against beta-lactamase J. Med. Chem. 2008, 51, 2502– 2511Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXjtFSksb0%253D&md5=7736429aeafc6b17a5d0182e24df0dd4Comprehensive Mechanistic Analysis of Hits from High-Throughput and Docking Screens against β-LactamaseBabaoglu, Kerim; Simeonov, Anton; Irwin, John J.; Nelson, Michael E.; Feng, Brian; Thomas, Craig J.; Cancian, Laura; Costi, M. Paola; Maltby, David A.; Jadhav, Ajit; Inglese, James; Austin, Christopher P.; Shoichet, Brian K.Journal of Medicinal Chemistry (2008), 51 (8), 2502-2511CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)High-throughput screening (HTS) is widely used in drug discovery. Esp. for screens of unbiased libraries, false positives can dominate "hit lists"; their origins are much debated. Here we det. the mechanism of every active hit from a screen of 70,563 unbiased mols. against β-lactamase using quant. HTS (qHTS). Of the 1274 initial inhibitors, 95% were detergent-sensitive and were classified as aggregators. Among the 70 remaining were 25 potent, covalent-acting β-lactams. Mass spectra, counter-screens, and crystallog. identified 12 as promiscuous covalent inhibitors. The remaining 33 were either aggregators or irreproducible. No specific reversible inhibitors were found. We turned to mol. docking to prioritize mols. from the same library for testing at higher concns. Of 16 tested, 2 were modest inhibitors. Subsequent X-ray structures corresponded to the docking prediction. Analog synthesis improved affinity to 8 μM. These results suggest that it may be the phys. behavior of org. mols., not their reactivity, that accounts for most screening artifacts. Structure-based methods may prioritize weak-but-novel chemotypes in unbiased library screens.
- 29Powers, R. A.; Morandi, F.; Shoichet, B. K. Structure-based discovery of a novel, noncovalent inhibitor of AmpC beta-lactamase Structure 2002, 10, 1013– 1023Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XlsVyjsrk%253D&md5=5f539484d08191f0c65626493fc1b262Structure-Based Discovery of a Novel, Noncovalent Inhibitor of AmpC β-LactamasePowers, Rachel A.; Morandi, Federica; Shoichet, Brian K.Structure (Cambridge, MA, United States) (2002), 10 (7), 1013-1023CODEN: STRUE6; ISSN:0969-2126. (Cell Press)β-Lactamases are the most widespread resistance mechanisms to β-lactam antibiotics, and there is a pressing need for novel, non-β-lactam drugs. A database of over 200,000 compds. was docked to the active site of AmpC β-lactamase to identify potential inhibitors. Fifty-six compds. were tested, and three had Ki values of 650 μM or better. The best of these, 3-[(4-chloroanilino)sulfonyl]thiophene-2-carboxylic acid, was a competitive noncovalent inhibitor (Ki = 26 μM), which also reversed resistance to β-lactams in bacteria expressing AmpC. The structure of AmpC in complex with this compd. was detd. by x-ray crystallog. to 1.94 A and reveals that the inhibitor interacts with key active-site residues in sites targeted in the docking calcn. Indeed, the exptl. detd. conformation of the inhibitor closely resembles the prediction. The structure of the enzyme-inhibitor complex presents an opportunity to improve binding affinity in a novel series of inhibitors discovered by structure-based methods.
- 30Meng, E. C.; Shoichet, B. K.; Kuntz, I. D. Automated docking with grid-based energy evaluation J. Comput. Chem. 1992, 13, 505– 524Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38Xit1Omt7c%253D&md5=f472fffa0c9a61652b4c20e4dbbba69eAutomated docking with grid-based energy evaluationMeng, Elaine C.; Shoichet, Brian K.; Kuntz, Irwin D.Journal of Computational Chemistry (1992), 13 (4), 505-24CODEN: JCCHDD; ISSN:0192-8651.The ability to generate feasible binding orientations of a small mol. within a site of known structure is important for ligand design. The authors present a method that combines a rapid, geometric docking algorithm with the evaluation of mol. mechanics interaction energies. The computational costs of evaluation are minimal because the authors precalc. the receptor-dependent terms in the potential function at points on a three-dimensional grid. In four test cases where the components of crystallog. detd. complexes are redocked, the "force field" score correctly identifies the family of orientations closest to the exptl. binding geometry. Scoring functions that consider only steric factors or only electrostatic factors are less successful. The force field function will play an important role in efforts to search databases for potential lead compds.
- 31Shoichet, B. K.; Leach, A. R.; Kuntz, I. D. Ligand solvation in molecular docking Proteins: Struct., Funct., Genet. 1999, 34, 4– 16Google ScholarThere is no corresponding record for this reference.
- 32Wei, B. Q. Q.; Baase, W. A.; Weaver, L. H.; Matthews, B. W.; Shoichet, B. K. A model binding site for testing scoring functions in molecular docking J. Mol. Biol. 2002, 322, 339– 355Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xms1Wqsrs%253D&md5=040e009b33f125ab4abbb322cf32d2c9A Model Binding Site for Testing Scoring Functions in Molecular DockingWei, Binqing Q.; Baase, Walter A.; Weaver, Larry H.; Matthews, Brian W.; Shoichet, Brian K.Journal of Molecular Biology (2002), 322 (2), 339-355CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Science Ltd.)Prediction of interaction energies between ligands and their receptors remains a major challenge for structure-based inhibitor discovery. Much effort has been devoted to developing scoring schemes that can successfully rank the affinities of a diverse set of possible ligands to a binding site for which the structure is known. To test these scoring functions, well-characterized exptl. systems can be very useful. Here, mutation-created binding sites in T4 lysozyme were used to investigate how the quality of at. charges and solvation energies affects mol. docking. At. charges and solvation energies were calcd. for 172,118 mols. in the Available Chems. Directory using a semi-empirical quantum mech. approach by the program AMSOL. The database was first screened against the apolar cavity site created by the mutation Leu99Ala (L99A). Compared to the electronegativity-based charges that are widely used, the new charges and desolvation energies improved ranking of known apolar ligands, and better distinguished them from more polar isosteres that are not obsd. to bind. To investigate whether the new charges had predictive value, the non-polar residue Met102, which forms part of the binding site, was changed to the polar residue glutamine. The structure of the resulting Leu99 Ala and Met102 Gln double mutant of T4 lysozyme (L99A/M102Q) was detd. and the docking calcn. was repeated for the new site. Seven representative polar mols. that preferentially docked to the polar vs. the apolar binding site were tested exptl. All seven bind to the polar cavity (L99A/M102Q) but do not detectably bind to the apolar cavity (L99A). Five ligand-bound structures of L99A/M102Q were detd. by X-ray crystallog. Docking predictions corresponded to the crystallog. results to within 0.4 A RMSD. Improved treatment of partial at. charges and desolvation energies in database docking appears feasible and leads to better distinction of true ligands. Simple model binding sites, such as L99A and its more polar variants, may find broad use in the development and testing of docking algorithms.
- 33Irwin, J. J.; Shoichet, B. K. ZINC—a free database of commercially available compounds for virtual screening J. Chem. Inf. Model. 2005, 45, 177– 182Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVOjt77J&md5=e3892b7dc8608b17a3e63541a5ed60e6ZINC - A Free Database of Commercially Available Compounds for Virtual ScreeningIrwin, John J.; Shoichet, Brian K.Journal of Chemical Information and Computer Sciences (2005), 45 (1), 177-182CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)A crit. barrier to entry into structure-based virtual screening is the lack of a suitable, easy to access database of purchasable compds. We have therefore prepd. a library of 727 842 mols., each with 3D structure, using catalogs of compds. from vendors (the size of this library continues to grow). The mols. have been assigned biol. relevant protonation states and are annotated with properties such as mol. wt., calcd. LogP, and no. of rotatable bonds. Each mol. in the library contains vendor and purchasing information and is ready for docking using a no. of popular docking programs. Within certain limits, the mols. are prepd. in multiple protonation states and multiple tautomeric forms. In one format, multiple conformations are available for the mols. This database is available for free download (http://zinc.docking.org) in several common file formats including SMILES, mol2, 3D SDF, and DOCK flexibase format. A Web-based query tool incorporating a mol. drawing interface enables the database to be searched and browsed and subsets to be created. Users can process their own mols. by uploading them to a server. Our hope is that this database will bring virtual screening libraries to a wide community of structural biologists and medicinal chemists.
- 34Bostrom, J.; Greenwood, J. R.; Gottfries, J. Assessing the performance of OMEGA with respect to retrieving bioactive conformations J. Mol. Graphics Modell. 2003, 21, 449– 462Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXkvFarsQ%253D%253D&md5=6c690a35f8792114a3da7b2afb485140Assessing the performance of OMEGA with respect to retrieving bioactive conformationsBostrom, Jonas; Greenwood, Jeremy R.; Gottfries, JohanJournal of Molecular Graphics & Modelling (2003), 21 (5), 449-462CODEN: JMGMFI; ISSN:1093-3263. (Elsevier Science Inc.)OMEGA is a rule-based program which rapidly generates conformational ensembles of small mols. We have varied the parameters which control the nature of the ensembles generated by OMEGA in a statistical fashion (D-optimal) with the aim of increasing the probability of generating bioactive conformations. Thirty-six drug-like ligands from different ligand-protein complexes detd. by high-resoln. (≤ 2.0 Å) x-ray crystallog. have been analyzed. Statistically significant models (Q2 ≥ 0.75) confirm that one can increase the performance of OMEGA by modifying the parameters. Twenty-eight of the bioactive conformations were retrieved when using a low-energy cut-off (5 kcal/mol), a low RMSD value (0.6 Å) for duplicate removal, and a max. of 1000 output conformations. All of those that were not retrieved had eight or more rotatable bonds. The duplicate removal parameter was found to have the largest impact on retrieval of bioactive conformations, and the max. no. of conformations also affected the results considerably. The input conformation was found to influence the results largely because certain bond angles can prevent the bioactive conformation from being generated as a low-energy conformation. Pre-optimizing the input structures with MMFF94s improved the results significantly. We also investigated the performance of OMEGA in connection with database searching. The shape-matching program Rapid Overlay of Chem. Structures (ROCS) was used as search tool. Two multi-conformational databases were built from the MDDR database plus the 36 compds.; one large (max. 1000 conformations/mol) and one small (max. 100 conformations/mol). Both databases provided satisfactory results in terms of retrieval. ROCS was able to rank 35 out of 36 x-ray structures among the top 500 hits from the large database.
- 35Chambers, C. C.; Hawkins, G. D.; Cramer, C. J.; Truhlar, D. G. Model for aqueous solvation based on class IV atomic charges and first solvation shell effects J. Phys. Chem. 1996, 100, 16385– 16398Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XlsFCqurw%253D&md5=2ff8cd5c0e9d4f37070c0cdf3829fe7eModel for Aqueous Solvation Based on Class IV Atomic Charges and First Solvation Shell EffectsChambers, Candee C.; Cramer, Christopher J.; Truhlar, Donald G.Journal of Physical Chemistry (1996), 100 (40), 16385-16398CODEN: JPCHAX; ISSN:0022-3654. (American Chemical Society)A new set of geometry-based functional forms is presented for parameterizing effective Coulomb radii and at. surface tensions of org. solutes in water. In particular, the radii and surface tensions depend in some cases on distances to nearby atoms. Combining the surface tensions with electrostatic effects included in a Fock operator by the generalized Born model enables one to calc. free energies of solvation, and exptl. free energies of solvation are used to parametrize the theory for water. At. charges are obtained by both the AM1-CM1A and PM3-CM1P class IV charge models, which yield similar results, and hence the same radii and surface tensions are used with both charge models. The authors considered 215 neutral solutes contg. H, C, N, O, F, S, Cl, Br, and I and encompassing a very wide variety of org. functional groups, and a mean unsigned error in the free energy of hydration of 0.50 kcal/mol using CM1A charges and 0.48 kcal/mol using CM1P charges was obtained. The predicted solvation energies for 12 cationic and 22 anionic solutes have mean unsigned deviations from expt. of 4.6 and 4.8 kcal/mol for models based on AM1 and PM3, resp.
- 36Li, J. B.; Zhu, T. H.; Cramer, C. J.; Truhlar, D. G. New class IV charge model for extracting accurate partial charges from wave functions J. Phys. Chem. A 1998, 102, 1820– 1831Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXhtVegtr0%253D&md5=4fe859556188d73500054bb1ebdb0843New Class IV Charge Model for Extracting Accurate Partial Charges from Wave FunctionsLi, Jiabo; Zhu, Tianhai; Cramer, Christopher J.; Truhlar, Donald G.Journal of Physical Chemistry A (1998), 102 (10), 1820-1831CODEN: JPCAFH; ISSN:1089-5639. (American Chemical Society)We propose a new formalism, charge model 2 (CM2), to obtain accurate partial at. charges from a population anal. of wave functions by a parametrized mapping procedure, so that the resulting charges reproduce highly accurate charge-dependent observables. The new method, which produces class IV charges, is illustrated by developing CM2 mappings of Lowdin charges obtained from semiempirical and ab initio Hartree-Fock theory and d. functional theory, in particular AM1, PM3, HF/MIDI!, HF/6-31G*, HF/6-31+G*, BPW91/MIDI!, BPW91/6-31G*, B3LYP/MIDI!, and BPW91/DZVP calcns. The CM2 partial charges reproduce exptl. dipole moments with root-mean-square errors that are typically a factor of 7 better than dipole moments computed from Mulliken population anal., a factor of 3 better than dipole moments computed by Lowdin anal., and even a factor of 2 better than dipole moments computed from the continuous electron d. At the HF/6-31G* and B3LYP/MIDI! levels, the new charge model yields root-mean-square errors of 0.19 and 0.18 D, resp., for the dipole moments of a set of 211 polar mols. contg. a diverse range of structures and org. functional groups and the elements H, C, N, O, F, Si, P, S, Cl, Br, and I. A comparison shows that the new charge model predicts dipole moments more accurately than MP2/cc-pVDZ calcns., which are considerably more expensive. The quality of the results is similarly good for electrostatic potentials and for the other parametrizations as well.
- 37Weiner, S. J.; Kollman, P. A.; Nguyen, D. T.; Case, D. A. An all atom force-field for simulations of proteins and nucleic-acids J. Comput. Chem. 1986, 7, 230– 252Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XhvVarsLY%253D&md5=33f0620f30d45081f9383240f4dacd73An all atom force field for simulations of proteins and nucleic acidsWeiner, Scott J.; Kollman, Peter A.; Nguyen, Dzung T.; Case, David A.Journal of Computational Chemistry (1986), 7 (2), 230-52CODEN: JCCHDD; ISSN:0192-8651.An all-atom potential energy function for the simulation of proteins and nucleic acids is presented. This work is an extension of the CH united atom function recently presented by S. J. Weiner et al. (1984). The parameters of this function are based on calcns. on ethane, propane, n-butane, di-Me ether, Me Et ether, THF, imidazole, indole, deoxyadenosine, base-paired dinucleoside phosphates, adenine, guanine, uracil, cytosine, thymine, insulin, and myoglobin. These parameters were also used to carry out the 1st general vibrational anal. of all 5 nucleic acid bases with a mol. mechanics potential approach.
- 38http://accelrys.com/products/scitegic/.Google ScholarThere is no corresponding record for this reference.
- 39Olah, M.; Mracec, M.; Ostopovici, L.; Rad, R.; Bora, A.; Hadaruga, N.; Olah, I.; Banda, M.; Simon, Z.; Mracec, M.; Oprea, T. I. WOMBAT: World of Molecular Bioactivity. In Chemoinformatics in Drug Discovery; Oprea, T. I., Ed.; Wiley-VCH: Weinheim, Germany, 2005; pp 221− 239.Google ScholarThere is no corresponding record for this reference.
- 40http://www.ebi.ac.uk/chembl.Google ScholarThere is no corresponding record for this reference.
- 41Keiser, M. J.; Roth, B. L.; Armbruster, B. N.; Ernsberger, P.; Irwin, J. J.; Shoichet, B. K. Relating protein pharmacology by ligand chemistry Nat. Biotechnol. 2007, 25, 197– 206Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlOrsLo%253D&md5=1b7373d52563fca5fe1e893d85f70573Relating protein pharmacology by ligand chemistryKeiser, Michael J.; Roth, Bryan L.; Armbruster, Blaine N.; Ernsberger, Paul; Irwin, John J.; Shoichet, Brian K.Nature Biotechnology (2007), 25 (2), 197-206CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The identification of protein function based on biol. information is an area of intense research. Here the authors consider a complementary technique that quant. groups and relates proteins based on the chem. similarity of their ligands. The authors began with 65,000 ligands annotated into sets for hundreds of drug targets. The similarity score between each set was calcd. using ligand topol. A statistical model was developed to rank the significance of the resulting similarity scores, which are expressed as a min. spanning tree to map the sets together. Although these maps are connected solely by chem. similarity, biol. sensible clusters nevertheless emerged. Links among unexpected targets also emerged, among them that methadone, emetine and loperamide (Imodium) may antagonize muscarinic M3, α2 adrenergic and neurokinin NK2 receptors, resp. These predictions were subsequently confirmed exptl. Relating receptors by ligand chem. organizes biol. to reveal unexpected relationships that may be assayed using the ligands themselves.
- 42Tondi, D.; Morandi, F.; Bonnet, R.; Costi, M. P.; Shoichet, B. K. Structure-based optimization of a non-beta-lactam lead results in inhibitors that do not up-regulate beta-lactamase expression in cell culture J. Am. Chem. Soc. 2005, 127, 4632– 4639Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXit1Krsbk%253D&md5=ddad35ac3e6fb96cf65e12cd807f271cStructure-Based Optimization of a Non-β-lactam Lead Results in Inhibitors That Do Not Up-Regulate β-Lactamase Expression in Cell CultureTondi, Donatella; Morandi, Federica; Bonnet, Richard; Costi, M. Paola; Shoichet, Brian K.Journal of the American Chemical Society (2005), 127 (13), 4632-4639CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Bacterial expression of β-lactamases is the most widespread resistance mechanism to β-lactam antibiotics, such as penicillins and cephalosporins. There is a pressing need for novel, non-β-lactam inhibitors of these enzymes. One previously discovered novel inhibitor of the β-lactamase AmpC (I) has several favorable properties: it is chem. dissimilar to β-lactams and is a noncovalent, competitive inhibitor of the enzyme. However, at 26 μM its activity is modest. Using the x-ray structure of the AmpC/I complex as a template, 14 analogs were designed and synthesized. The most active of these, (II), had a Ki of 1 μM, 26-fold better than the lead. To understand the origins of this improved activity, the structures of AmpC in complex with compd. II and an analog were detd. by x-ray crystallog. to 1.97 and 1.96 Å, resp. II was active in cell culture, reversing resistance to the third generation cephalosporin ceftazidime in bacterial pathogens expressing AmpC. In contrast to β-lactam-based inhibitors clavulanate and cefoxitin, compd. II did not up-regulate β-lactamase expression in cell culture but simply inhibited the enzyme expressed by the resistant bacteria. Its escape from this resistance mechanism derives from its dissimilarity to β-lactam antibiotics.
- 43Jarvis, M. F.; Schulz, R.; Hutchison, A. J.; Do, U. H.; Sills, M. A.; Williams, M. [H-3] Cgs-21680, a selective A2 adenosine receptor agonist directly labels A2-receptors in rat-brain J. Pharmacol. Exp. Ther. 1989, 251, 888– 893Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXhtFamur4%253D&md5=fc1961da1f86a5592fa6142878952fa9[3H]CGS 21680, a selective A2 adenosine receptor agonist directly labels A2 receptors in rat brainJarvis, Michael F.; Schulz, Rainer; Hutchison, Alan J.; Do, Un Hoi; Sills, Matthew A.; Williams, MichaelJournal of Pharmacology and Experimental Therapeutics (1989), 251 (3), 888-93CODEN: JPETAB; ISSN:0022-3565.Characterization of the adenosine A2 receptor has been limited due to the lack of available ligands which have high affinity and selectivity for this adenosine receptor subtype. In the present study, the binding of a highly A2-selective agonist radioligand, [3H]CGS 21680 (I) is described. [3H]CGS 21680 specific binding to rat striatal membranes was saturable, reversible, and dependent on protein concn. Satn. studies revealed that [3H]CGS 21680 bound with high affinity (Kd =15.5 nM) and limited capacity (apparent Bmax = 375 fmol/mg protein) to a single class of recognition sites. Ests. of ligand affinity (16 nM) detd. from assocn. and dissocn. kinetic expts. were in close agreement with the results from the satn. studies. [3H]CGS 21680 binding was greatest in striatal membranes with negligible specific binding obtained in rat cortical membranes. Adenosine agonists ligands competed for the binding of 5 nM [3H]CGS 21680 to striatal membranes with the following order of activity; CGS 21680 = 5'-N-ethylcarboxamidoadenosine > 2-phenylaminoadenosine (CV-1808) ; 5'-N-methylcarboxamidoadenosine = 2-chloroadenosine > R-phenylisopropyladenosine > N6-cyclohexyladenosine > N6-cyclopentyltheophylline > S-phenylisopropyladenosine. The nonxanthine adenosine antagonist, CGS 15943A, was the most active compd. in inhibiting the binding of [3H]CGS 21680. Other adenosine antagonists inhibited binding in the following order; xanthine amine congener = (1,3-dipropyl-8-(2-amino-4-chloro)phenylxanthine > 1,3-dipropyl-8-cyclopentylxanthine > 1,3-diethyl-8-phenylxanthine > 8-phenyltheophylline > 8-cyclopentyltheophylline = xanthine carboxylic acid congener > 8-parasulfophenyltheophylline > theophylline > caffeine. The pharmacol. profile of both adenosine agonist and antagonist compds. to compete for the binding of [3H]CGS 21680 was consistent with a selective interaction at the high affinity adenosine A2 receptor. A high pos. correlation was obsd. between the pharmacol. profile of adenosine ligands to inhibit the binding of [3H]CGS 21680 and the selective binding of [3H]NECA (+50 nM CPA) to high affinity A2 receptors. However, some differences between these assays were found for compds. which have moderate affinity and nonselective actions at both the A1 and A2 adenosine receptor subtypes. Unlike data obtained with nonselective adenosine ligands, the present results indicate that [3H]CGS 21680 directly labels the high affinity A2 receptor in rat brain without the need to block binding activity at the A1 receptor. The high degree of selectivity (>170-fold) and high affinity of [3H]CGS 21680 make this the current ligand of choice for the in vitro characterization of high affinity A2 receptors.
- 44Klotz, K. N.; Lohse, M. J.; Schwabe, U.; Cristalli, G.; Vittori, S.; Grifantini, M. 2-Chloro-N-6-[H-3]cyclopentyladenosine ([H-3]Ccpa), a high-affinity agonist radioligand for A1 adenosine receptors Naunyn-Schmiedeberg's Arch. Pharmacol. 1989, 340, 679– 683Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXhtFGhsL8%253D&md5=d44acdedb7aa0c3b3c6f7d79666ae1392-Chloro-N6-[3H]cyclopentyladenosine ([3H]CPPA) - a high affinity agonist radioligand for A1 adenosine receptorsKlotz, Karl Norbert; Lohse, Martin J.; Schwabe, Ulrich; Cristalli, Gloria; Vittori, Sauro; Grifantini, MarioNaunyn-Schmiedeberg's Archives of Pharmacology (1989), 340 (6), 679-83CODEN: NSAPCC; ISSN:0028-1298.The tritiated analog of 2-chloro-N6-cyclopentyladenosine (CCPA) (I), an adenosine deriv. with subnanomolar affinity and a 10,000-fold selectivity for A1 adenosine receptors, has been examd. as a new agonist radioligand. [3H]CCPA was prepd. with a specific radioactivity of 1.58 TBq/mmol (43 Ci/mmol) and bound in a reversible manner to A1 receptors from rat brain membranes with a high affinity KD value of 0.2 nmol/L. In the presence of GTP, a KDvalue of 13 nmol/L was detd. for the low affinity state for agonist binding. Competition of several adenosine receptor agonists and antagonists for [3H]CCPA binding to rat brain membranes confirmed binding to an A1 receptor. Solubilized A1 receptors bound [3H]CCPA with similar affinity for the high affinity state. At solubilized receptors a reduced assocn. rate was obsd. in the presence of MgCl2, as has been shown for the agonist [3H]N6-phenylisopropyladenosine ([3H]PIA). [3H]CCPA was also used for detection of A1 receptors in rat cardiomyocyte membranes, a tissue with a very low receptor d. Kd-Value of 0.4 nmol-L and a Bmax-value of 16 fmol-platelet membranes, no specific binding of [3H]CCPA was measured at concns. up to 400 nmol/L, indicating that A2 receptors did not bind [3H]CCPA. Based on the subnanomolar affinity and the high selectivity for A1 receptors, [3H]CCPA proved to be a useful agonist radioligand for characterization of A1 adenosine receptors also in tissues with very low receptor d.
- 45Olah, M. E.; Gallorodriguez, C.; Jacobson, K. A.; Stiles, G. L. I-125 4-aminobenzyl-5′-N-methylcarboxamidoadenosine, a high-affinity radioligand for the rat a(3) adenosine receptor Mol. Pharmacol. 1994, 45, 978– 982Google ScholarThere is no corresponding record for this reference.
- 46Englert, M.; Quitterer, U.; Klotz, K. N. Effector coupling of stably transfected human A(3) adenosine receptors in CHO cells Biochem. Pharmacol. 2002, 64, 61– 65Google ScholarThere is no corresponding record for this reference.
- 47Jacobson, K. A.; Park, K. S.; Jiang, J. L.; Kim, Y. C.; Olah, M. E.; Stiles, G. L.; Ji, X. D. Pharmacological characterization of novel A(3) adenosine receptor-selective antagonists Neuropharmacology 1997, 36, 1157– 1165Google ScholarThere is no corresponding record for this reference.
- 48Nordstedt, C.; Fredholm, B. B. A modification of a protein-binding method for rapid quantification of camp in cell-culture supernatants and body-fluid Anal. Biochem. 1990, 189, 231– 234Google ScholarThere is no corresponding record for this reference.
- 49Post, S. R.; Ostrom, R. S.; Insel, P. A. Biochemical methods for detection and measurement of cyclic AMP and adenylyl cyclase activity Methods Mol. Biol. 2000, 126, 363– 374Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXjslWmuw%253D%253D&md5=0c9a58e800650d2f7d0ea512017472a0Biochemical methods for detection and measurement of cyclic AMP and adenylyl cyclase activityPost, Steven R.; Ostrom, Rennolds S.; Insel, Paul A.Methods in Molecular Biology (Totowa, New Jersey) (2000), 126 (Adrenergic Receptor Protocols), 363-374CODEN: MMBIED; ISSN:1064-3745. (Humana Press Inc.)Several assays are described and detailed for the characterization and anal. of cAMP and adenylyl cyclase.
- 50Bradford, M. M. Rapid and sensitive method for quantitation of microgram quantities of protein utilizing principle of protein−dye binding Anal. Biochem. 1976, 72, 248– 254Google ScholarThere is no corresponding record for this reference.
- 51McGovern, S. L.; Helfand, B. T.; Feng, B.; Shoichet, B. K. A specific mechanism of nonspecific inhibition J. Med. Chem. 2003, 46, 4265– 4272Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXmsFamsbs%253D&md5=cd0fd6bf0a64ceb7d7219dae2406dd21A Specific Mechanism of Nonspecific InhibitionMcGovern, Susan L.; Helfand, Brian T.; Feng, Brian; Shoichet, Brian K.Journal of Medicinal Chemistry (2003), 46 (20), 4265-4272CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Promiscuous small mols. plague screening libraries and hit lists. Previous work has found that several nonspecific compds. form submicrometer aggregates, and it has been suggested that this aggregate species is responsible for the inhibition of many different enzymes. It is not understood how aggregates inhibit their targets. To address this question, biophys., kinetic, and microscopy methods were used to study the interaction of promiscuous, aggregate-forming inhibitors with model proteins. By use of centrifugation and gel electrophoresis, aggregates and protein were found to directly interact. This is consistent with a subsequent observation from confocal fluorescence microscopy that aggregates conc. green fluorescent protein. β-Lactamase mutants with increased or decreased thermodn. stability relative to wild-type enzyme were equally inhibited by an aggregate-forming compd., suggesting that denaturation by unfolding was not the primary mechanism of interaction. Instead, visualization by electron microscopy revealed that enzyme assocs. with the surface of inhibitor aggregates. This assocn. could be reversed or prevented by the addn. of Triton X-100. These observations suggest that the aggregates formed by promiscuous compds. reversibly sequester enzyme, resulting in apparent inhibition. They also suggest a simple method to identify or reverse the action of aggregate-based inhibitors, which appear to be widespread.
- 52Kim, J. H.; Wess, J.; Vanrhee, A. M.; Schoneberg, T.; Jacobson, K. A. Site-directed mutagenesis identifies residues involved in ligand recognition in the human a(2a) adenosine receptor J. Biol. Chem. 1995, 270, 13987– 13997Google ScholarThere is no corresponding record for this reference.
- 53Webb, T. R.; Lvovskiy, D.; Kim, S. A.; Ji, X. D.; Melman, N.; Linden, J.; Jacobson, K. A. Quinazolines as adenosine receptor antagonists: SAR and selectivity for A(2B) receptors Bioorg. Med. Chem. 2003, 11, 77– 85Google ScholarThere is no corresponding record for this reference.
- 54Hert, J.; Irwin, J. J.; Laggner, C.; Keiser, M. J.; Shoichet, B. K. Quantifying biogenic bias in screening libraries Nat. Chem. Biol. 2009, 5, 479– 483Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXms1aksbg%253D&md5=67a631cb210e4cf7aa56190f7e2d7e2bQuantifying biogenic bias in screening librariesHert, Jerome; Irwin, John J.; Laggner, Christian; Keiser, Michael J.; Shoichet, Brian K.Nature Chemical Biology (2009), 5 (7), 479-483CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)In lead discovery, libraries of 106 mols. are screened for biol. activity. Given the over 1060 drug-like mols. thought possible, such screens might never succeed. The fact that they do, even occasionally, implies a biased selection of library mols. We have developed a method to quantify the bias in screening libraries toward biogenic mols. With this approach, we consider what is missing from screening libraries and how they can be optimized.
- 55Soelaiman, S.; Wei, B. Q.; Bergson, P.; Lee, Y. S.; Shen, Y.; Mrksich, M.; Shoichet, B. K.; Tang, W. J. Structure-based inhibitor discovery against adenylyl cyclase toxins from pathogenic bacteria that cause anthrax and whooping cough J. Biol. Chem. 2003, 278, 25990– 25997Google ScholarThere is no corresponding record for this reference.
- 56Katritch, V.; Jaakola, V. P.; Lane, J. R.; Lin, J.; Ijzerman, A. P.; Yeager, M.; Kufareva, I.; Stevens, R. C.; Abagyan, R. Structure-based discovery of novel chemotypes for adenosine A(2A) receptor antagonists J. Med. Chem. 2010, 53, 1799– 1809Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptlKqsw%253D%253D&md5=5a54df43f6edd20d83e7e5942e2f9811Structure-Based Discovery of Novel Chemotypes for Adenosine A2A Receptor AntagonistsKatritch, Vsevolod; Jaakola, Veli-Pekka; Lane, J. Robert; Lin, Judy; IJzerman, Adriaan P.; Yeager, Mark; Kufareva, Irina; Stevens, Raymond C.; Abagyan, RubenJournal of Medicinal Chemistry (2010), 53 (4), 1799-1809CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent progress in crystallog. of G-protein coupled receptors opens an unprecedented venue for structure-based GPCR drug discovery. To test efficiency of the structure-based approach, we performed mol. docking and virtual ligand screening (VLS) of more than 4 million com. available "drug-like" and "lead-like" compds. against the A2AAR 2.6 Å resoln. crystal structure. Out of 56 high ranking compds. tested in A2AAR binding assays, 23 showed affinities under 10 μM, 11 of those had sub-μM affinities and two compds. had affinities under 60 nM. The identified hits represent at least 9 different chem. scaffolds and are characterized by very high ligand efficiency (0.3-0.5 kcal/mol per heavy atom). Significant A2AAR antagonist activities were confirmed for 10 out of 13 ligands tested in functional assays. High success rate, novelty, and diversity of the chem. scaffolds and strong ligand efficiency of the A2AAR antagonists identified in this study suggest practical applicability of receptor-based VLS in GPCR drug discovery.
Supporting Information
Supporting Information
ARTICLE SECTIONSTable S1 of structures of the 500 top-ranking molecules from the docking screen. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.